1
|
Edelmann D, Terzer T, Horak P, Schlenk R, Benner A. The Progression-Free-Survival Ratio in Molecularly Aided Tumor Trials: A Critical Examination of Current Practice and Suggestions for Alternative Methods. Biom J 2025; 67:e70028. [PMID: 39692541 DOI: 10.1002/bimj.70028] [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: 01/16/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 12/19/2024]
Abstract
The progression-free-survival ratio is a popular endpoint in oncology trials, which is frequently applied to evaluate the efficacy of molecularly targeted treatments in late-stage patients. Using elementary calculations and simulations, numerous shortcomings of the current methodology are pointed out. As a remedy to these shortcomings, an alternative methodology is proposed, using a marginal Cox model or a marginal accelerated failure time model for clustered time-to-event data. Using comprehensive simulations, it is shown that this methodology outperforms existing methods in settings where the intrapatient correlation is low to moderate. The performance of the model is further demonstrated in a real data example from a molecularly aided tumor trial. Sample size considerations are discussed.
Collapse
Affiliation(s)
- Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Terzer
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Horak
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Richard Schlenk
- Department of Internal Medicine V and Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
- NCT Trial Center, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
2
|
Bisht VS, Kumar D, Najar MA, Giri K, Kaur J, Prasad TSK, Ambatipudi K. Drug response-based precision therapeutic selection for tamoxifen-resistant triple-positive breast cancer. J Proteomics 2025; 310:105319. [PMID: 39299547 DOI: 10.1016/j.jprot.2024.105319] [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: 06/20/2024] [Revised: 09/15/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Breast cancer adaptability to the drug environment reduces the chemotherapeutic response and facilitates acquired drug resistance. Cancer-specific therapeutics can be more effective against advanced-stage cancer than standard chemotherapeutics. To extend the paradigm of cancer-specific therapeutics, clinically relevant acquired tamoxifen-resistant MCF-7 proteome was deconstructed to identify possible druggable targets (N = 150). Twenty-eight drug inhibitors were used against identified druggable targets to suppress non-resistant (NC) and resistant cells (RC). First, selected drugs were screened using growth-inhibitory response against NC and RC. Seven drugs were shortlisted for their time-dependent (10-12 days) cytotoxic effect and further narrowed to three effective drugs (e.g., cisplatin, doxorubicin, and hydroxychloroquine). The growth-suppressive effectiveness of selected drugs was validated in the complex spheroid model (progressive and regressive). In the progressive model, doxorubicin (RC: 83.64 %, NC: 54.81 %), followed by cisplatin (RC: 76.66 %, NC: 68.94 %) and hydroxychloroquine (RC: 68.70 %, NC: 61.78 %) showed a significant growth-suppressive effect. However, in fully grown regressive spheroid, after 4th drug treatment, cisplatin significantly suppressed RC (84.79 %) and NC (40.21 %), while doxorubicin and hydroxychloroquine significantly suppressed only RC (76.09 and 76.34 %). Our in-depth investigation effectively integrated the expression data with the cancer-specific therapeutic investigation. Furthermore, our three-step sequential drug-screening approach unbiasedly identified cisplatin, doxorubicin, and hydroxychloroquine as an efficacious drug to target heterogeneous cancer cell populations. SIGNIFICANCE STATEMENT: Hormonal-positive BC grows slowly, and hormonal-inhibitors effectively suppress the oncogenesis. However, development of drug-resistance not only reduces the drug-response but also increases the chance of BC aggressiveness. Further, alternative chemotherapeutics are widely used to control advanced-stage BC. In contrast, we hypothesized that, compared to standard chemotherapeutics, cancer-specific drugs can be more effective against resistant-cancer. Although cancer-specific treatment identification is an uphill battle, our work shows proteome data can be used for drug selection. We identified multiple druggable targets and, using ex-vivo methods narrowed multiple drugs to disease-condition-specific therapeutics. We consider that our investigation successfully interconnected the expression data with the functional disease-specific therapeutic investigation and selected drugs can be used for effective resistant treatment with higher therapeutic response.
Collapse
Affiliation(s)
- Vinod S Bisht
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Deepak Kumar
- Department of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Kuldeep Giri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Jaismeen Kaur
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | | | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| |
Collapse
|
3
|
Ben Cohen G, Yaacov A, Ben Zvi Y, Loutati R, Lishinsky N, Landau J, Hope T, Popovzter A, Rosenberg S. Graph convolution networks model identifies and quantifies gene and cancer specific transcriptome signatures of cancer driver events. Comput Biol Med 2024; 185:109491. [PMID: 39700860 DOI: 10.1016/j.compbiomed.2024.109491] [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: 05/20/2024] [Revised: 08/24/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The identification and drug targeting of cancer causing (driver) genetic alterations has seen immense improvement in recent years, with many new targeted therapies developed. However, identifying, prioritizing, and treating genetic alterations is insufficient for most cancer patients. Current clinical practices rely mainly on DNA level mutational analyses, which in many cases fail to identify treatable driver events. Arguably, signal strength may determine cell fate more than the mutational status that initiated it. The use of transcriptomics, a complex and highly informative representation of cellular and tumor state, had been suggested to enhance diagnostics and treatment successes. A gene-expression based model trained over known genetic alterations could improve identification and quantification of cancer related biological aberrations' signal strength. METHODS We present STAMP (Signatures in Transcriptome Associated with Mutated Protein), a Graph Convolution Networks (GCN) based framework for the identification of gene expression signatures related to cancer driver events. STAMP was trained to identify the p53 dysfunction of cancer samples from gene expression, utilizing comprehensive curated graph structures of gene interactions. Predictions were modified for generating a quantitative score to rank the severity of a driver event in each sample. STAMP was then extended to almost 300 tumor type-specific predictive models for important cancer genes/pathways, by training to identify well-established driver events' annotations from the literature. RESULTS STAMP achieved very high AUC on unseen data across several tumor types and on an independent cohort. The framework was validated on p53 related genetic and clinical characteristics, including the effect of Variants of Unknown Significance, and showed strong correlation with protein function. For genes and tumor types where targeted therapy is available, STAMP showed correlation with drugs sensitivity (IC50) in an independent cell line database. It managed to stratify drug effect on samples with similar mutational profiles. STAMP was validated for drug-response prediction in clinical patients' cohorts, improving over a state-of-the-art method and suggesting potential biomarkers for cancer treatments. CONCLUSIONS The STAMP models provide a learning framework that successfully identifies and quantifies driver events' signal strength, showing utility in portraying the molecular landscape of tumors based on transcriptomics. Importantly, STAMP manifested the ability to improve targeted therapy selection and hence can contribute to better treatment.
Collapse
Affiliation(s)
- Gil Ben Cohen
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Adar Yaacov
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Yishai Ben Zvi
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Ranel Loutati
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Natan Lishinsky
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Jakob Landau
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tom Hope
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Aron Popovzter
- Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shai Rosenberg
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
4
|
Stoppe C, Hill A, Christopher KB, Kristof AS. Toward Precision in Nutrition Therapy. Crit Care Med 2024:00003246-990000000-00430. [PMID: 39688452 DOI: 10.1097/ccm.0000000000006537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Precision in critical care nutrition is paramount, as it focuses nutrition interventions on those patients most likely to benefit, or those who might potentially be harmed. Critical care nutrition must therefore be tailored to individual metabolic needs as determined by factors that control the capacity for tissue homeostasis and anabolic responses. This ideally involves the accurate and timely assessment of macronutrient and micronutrient requirements, a careful evaluation of metabolic response mechanisms and the identification of circumstances that might interfere with the productive utilization of dietary substrates. Specific surrogate markers of metabolic response, such as blood glucose levels, urea levels, or nitrogen balance, might be used to evaluate the metabolic readiness for nutrition and to establish the timing, nature, and clinical effectiveness of nutrition interventions. Despite the pressing need to further develop more targeted approaches in critically ill patients, indices of immediate metabolic responses that correlate with favorable clinical outcomes are lacking. In addition, the development of precision approaches might address timely adjustments in protein, energy, or micronutrient supplementation based on evolving clinical conditions. Here, we review why precision tools are needed in critical care nutrition, our progress thus far, as well as promising approaches and technologies by which multidisciplinary healthcare teams can improve quality of care and clinical outcomes by individualizing nutrition interventions.
Collapse
Affiliation(s)
- Christian Stoppe
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
- Department of Cardiac Anesthesiology and Intensive Care Medicine, Charité Berlin, Berlin, Germany
| | - Aileen Hill
- Department of Anesthesiology and Department of Operative Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Kenneth B Christopher
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Arnold S Kristof
- Meakins-Christie Laboratories and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canda
- Departments of Critical Care and Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
Dyshlovoy SA, Paigin S, Afflerbach AK, Lobermeyer A, Werner S, Schüller U, Bokemeyer C, Schuh AH, Bergmann L, von Amsberg G, Joosse SA. Applications of Nanopore sequencing in precision cancer medicine. Int J Cancer 2024; 155:2129-2140. [PMID: 39031959 DOI: 10.1002/ijc.35100] [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: 03/09/2024] [Revised: 04/25/2024] [Accepted: 06/25/2024] [Indexed: 07/22/2024]
Abstract
Oxford Nanopore Technologies sequencing, also referred to as Nanopore sequencing, stands at the forefront of a revolution in clinical genetics, offering the potential for rapid, long read, and real-time DNA and RNA sequencing. This technology is currently making sequencing more accessible and affordable. In this comprehensive review, we explore its potential regarding precision cancer diagnostics and treatment. We encompass a critical analysis of clinical cases where Nanopore sequencing was successfully applied to identify point mutations, splice variants, gene fusions, epigenetic modifications, non-coding RNAs, and other pivotal biomarkers that defined subsequent treatment strategies. Additionally, we address the challenges of clinical applications of Nanopore sequencing and discuss the current efforts to overcome them.
Collapse
Affiliation(s)
- Sergey A Dyshlovoy
- Department of Oncology, Oxford Molecular Diagnostics Centre, University of Oxford, Level 4, John Radcliffe Hospital, Oxford, UK
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefanie Paigin
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Ann-Kristin Afflerbach
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annabelle Lobermeyer
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Paediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna H Schuh
- Department of Oncology, Oxford Molecular Diagnostics Centre, University of Oxford, Level 4, John Radcliffe Hospital, Oxford, UK
| | - Lina Bergmann
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunhild von Amsberg
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Weller J, Potthoff A, Zeyen T, Schaub C, Duffy C, Schneider M, Herrlinger U. Current status of precision oncology in adult glioblastoma. Mol Oncol 2024; 18:2927-2950. [PMID: 38899374 PMCID: PMC11619805 DOI: 10.1002/1878-0261.13678] [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: 11/16/2023] [Revised: 04/05/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The concept of precision oncology, the application of targeted drugs based on comprehensive molecular profiling, has revolutionized treatment strategies in oncology. This review summarizes the current status of precision oncology in glioblastoma (GBM), the most common and aggressive primary brain tumor in adults with a median survival below 2 years. Targeted treatments without prior target verification have consistently failed. Patients with BRAF V600E-mutated GBM benefit from BRAF/MEK-inhibition, whereas targeting EGFR alterations was unsuccessful due to poor tumor penetration, tumor cell heterogeneity, and pathway redundancies. Systematic screening for actionable molecular alterations resulted in low rates (< 10%) of targeted treatments. Efficacy was observed in one-third and currently appears to be limited to BRAF-, VEGFR-, and mTOR-directed treatments. Advancing precision oncology for GBM requires consideration of pathways instead of single alterations, new trial concepts enabling rapid and adaptive drug evaluation, a focus on drugs with sufficient bioavailability in the CNS, and the extension of target discovery and validation to the tumor microenvironment, tumor cell networks, and their interaction with immune cells and neurons.
Collapse
Affiliation(s)
- Johannes Weller
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Thomas Zeyen
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Christina Schaub
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Cathrina Duffy
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Ulrich Herrlinger
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| |
Collapse
|
7
|
Moll M, Hecker J, Platig J, Zhang J, Ghosh AJ, Pratte KA, Wang RS, Hill D, Konigsberg IR, Chiles JW, Hersh CP, Castaldi PJ, Glass K, Dy JG, Sin DD, Tal-Singer R, Mouded M, Rennard SI, Anderson GP, Kinney GL, Bowler RP, Curtis JL, McDonald ML, Silverman EK, Hobbs BD, Cho MH. Polygenic and transcriptional risk scores identify chronic obstructive pulmonary disease subtypes in the COPDGene and ECLIPSE cohort studies. EBioMedicine 2024; 110:105429. [PMID: 39509750 PMCID: PMC11570824 DOI: 10.1016/j.ebiom.2024.105429] [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: 05/21/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Genetic variants and gene expression predict risk of chronic obstructive pulmonary disease (COPD), but their effect on COPD heterogeneity is unclear. We aimed to define high-risk COPD subtypes using genetics (polygenic risk score, PRS) and blood gene expression (transcriptional risk score, TRS) and assess differences in clinical and molecular characteristics. METHODS We defined high-risk groups based on PRS and TRS quantiles by maximising differences in protein biomarkers in a COPDGene training set and identified these groups in COPDGene and ECLIPSE test sets. We tested multivariable associations of subgroups with clinical outcomes and compared protein-protein interaction networks and drug repurposing analyses between high-risk groups. FINDINGS We examined two high-risk omics-defined groups in non-overlapping test sets (n = 1133 NHW COPDGene, n = 299 African American (AA) COPDGene, n = 468 ECLIPSE). We defined "high activity" (low PRS, high TRS) and "severe risk" (high PRS, high TRS) subgroups. Participants in both subgroups had lower body-mass index (BMI), lower lung function, and alterations in metabolic, growth, and immune signalling processes compared to a low-risk (low PRS, low TRS) subgroup. "High activity" but not "severe risk" participants had greater prospective FEV1 decline (COPDGene: -51 mL/year; ECLIPSE: -40 mL/year) and proteomic profiles were enriched in gene sets perturbed by treatment with 5-lipoxygenase inhibitors and angiotensin-converting enzyme (ACE) inhibitors. INTERPRETATION Concomitant use of polygenic and transcriptional risk scores identified clinical and molecular heterogeneity amongst high-risk individuals. Proteomic and drug repurposing analysis identified subtype-specific enrichment for therapies and suggest prior drug repurposing failures may be explained by patient selection. FUNDING National Institutes of Health.
Collapse
Affiliation(s)
- Matthew Moll
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary, Critical Care, Sleep and Allergy, Veterans Affairs Boston Healthcare System, West Roxbury, MA, 02123, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - John Platig
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jingzhou Zhang
- The Pulmonary Center, Boston University Medical Center, Boston, MA 02118, USA
| | - Auyon J Ghosh
- Division of Pulmonary, Critical Care, and Sleep Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Katherine A Pratte
- Department of Biostatistics, National Jewish Health, Denver, CO, 80206, USA
| | - Rui-Sheng Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Davin Hill
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Iain R Konigsberg
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Joe W Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer G Dy
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, and Department of Medicine (Respiratory Division), University of British Columbia, Vancouver, BC, Canada
| | - Ruth Tal-Singer
- Global Allergy and Airways Patient Platform, Vienna, Austria
| | - Majd Mouded
- Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Stephen I Rennard
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Nebraska, Omaha, NE, 68198, USA
| | - Gary P Anderson
- Lung Health Research Centre, Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Victoria, Australia
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA; Medical Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, 48109, USA
| | - Merry-Lynn McDonald
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, 701, 19th Street S., LHRB 440, Birmingham, AL, 35233, USA; Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA
| | | | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Qi L, Li Z, Liu J, Chen X. Omics-Enhanced Nanomedicine for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409102. [PMID: 39473316 DOI: 10.1002/adma.202409102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Cancer nanomedicine has emerged as a promising approach to overcome the limitations of conventional cancer therapies, offering enhanced efficacy and safety in cancer management. However, the inherent heterogeneity of tumors presents increasing challenges for the application of cancer nanomedicine in both diagnosis and treatment. This heterogeneity necessitates the integration of advanced and high-throughput analytical techniques to tailor nanomedicine strategies to individual tumor profiles. Omics technologies, encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and more, provide unparalleled insights into the molecular and cellular mechanisms underlying cancer. By dissecting tumor heterogeneity across multiple levels, these technologies offer robust support for the development of personalized and precise cancer nanomedicine strategies. In this review, the principles, techniques, and applications of key omics technologies are summarized. Especially, the synergistic integration of omics and nanomedicine in cancer therapy is explored, focusing on enhanced diagnostic accuracy, optimized therapeutic strategies and the assessment of nanomedicine-mediated biological responses. Moreover, this review addresses current challenges and outlines future directions in the field of omics-enhanced nanomedicine. By offering valuable insights and guidance, this review aims to advance the integration of omics with nanomedicine, ultimately driving improved diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
| | - Jianping Liu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
9
|
Ababneh O, Nishizaki D, Kato S, Kurzrock R. Tumor necrosis factor superfamily signaling: life and death in cancer. Cancer Metastasis Rev 2024; 43:1137-1163. [PMID: 39363128 PMCID: PMC11554763 DOI: 10.1007/s10555-024-10206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 10/05/2024]
Abstract
Immune checkpoint inhibitors have shaped the landscape of cancer treatment. However, many patients either do not respond or suffer from later progression. Numerous proteins can control immune system activity, including multiple tumor necrosis factor (TNF) superfamily (TNFSF) and TNF receptor superfamily (TNFRSF) members; these proteins play a complex role in regulating cell survival and death, cellular differentiation, and immune system activity. Notably, TNFSF/TNFRSF molecules may display either pro-tumoral or anti-tumoral activity, or even both, depending on tumor type. Therefore, TNF is a prototype of an enigmatic two-faced mediator in oncogenesis. To date, multiple anti-TNF agents have been approved and/or included in guidelines for treating autoimmune disorders and immune-related toxicities after immune checkpoint blockade for cancer. A confirmed role for the TNFSF/TNFRSF members in treating cancer has proven more elusive. In this review, we highlight the cancer-relevant TNFSF/TNFRSF family members, focusing on the death domain-containing and co-stimulation members and their signaling pathways, as well as their complicated role in the life and death of cancer cells.
Collapse
Affiliation(s)
- Obada Ababneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium, Paris, France.
- Department of Medicine, MCW Cancer Center, Milwaukee, WI, USA.
- Department of Oncology, University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
10
|
Lutz S, D'Angelo A, Hammerl S, Schmutz M, Claus R, Fischer NM, Kramer F, Hammoud Z. Unveiling the Digital Evolution of Molecular Tumor Boards. Target Oncol 2024:10.1007/s11523-024-01109-1. [PMID: 39609355 DOI: 10.1007/s11523-024-01109-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/30/2024]
Abstract
Molecular tumor boards (MTB) are interdisciplinary conferences involving various experts discussing patients with advanced tumors, to derive individualized treatment suggestions based on molecular variants. These discussions involve using heterogeneous internal data, such as patient clinical data, but also external resources such as knowledge databases for annotations and search for relevant clinical studies. This imposes a certain level of complexity that requires huge effort to homogenize the data and use it in a speedy manner to reach the needed treatment. For this purpose, most institutions involving an MTB are heading toward automation and digitalization of the process, hence reducing manual work requiring human intervention and subsequently time in deriving personalized treatment suggestions. The tools are also used to better visualize the patient's data, which allows a refined overview for the board members. In this paper, we present the results of our thorough literature research about MTBs, their process, the most common knowledge bases, and tools used to support this decision-making process.
Collapse
Affiliation(s)
- Sebastian Lutz
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany.
| | - Alicia D'Angelo
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Sonja Hammerl
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Maximilian Schmutz
- Institute of Digital Medicine (IDM), Medical Faculty, University of Augsburg, Augsburg, Germany
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nina M Fischer
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Frank Kramer
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Zaynab Hammoud
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| |
Collapse
|
11
|
Navarro P, Beato C, Rodriguez-Moreno JF, Ruiz-Llorente S, Mielgo X, Pineda E, Navarro M, Bruixola G, Grazioso TP, Viudez A, Fuster J, Nogueron E, Mediano MD, Balaña C, Mendez C, Rodriguez RM, Del Barco Berron S, Gongora B, Carmona-Bayonas A, Garcia-Donas J. Prospective study of the real impact of fusion centered genomic assays in patient management in a national collaborative group: the GETHI-XX-16 study. Clin Transl Oncol 2024:10.1007/s12094-024-03745-5. [PMID: 39485597 DOI: 10.1007/s12094-024-03745-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/21/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE Precision medicine represents a paradigm shift in oncology. Access to genetic testing and targeted therapies is frequently limited. Assays based on DNA sequencing can miss druggable alterations. We aimed to determine the impact of a free access program to RNA tests in patient management. METHODS We designed a multicenter prospective observational study within the Spanish National Group for Translational Oncology and Rare and Orphan Tumors (GETTHI). Eligible patients were adults with solid cancers that had progressed on standard therapies. Tumor samples were analyzed using two RNA sequencing assays (Trailblaze PharosTM and Archer FusionPlex Solid TumorTM). A central committee evaluated the actionability of genetic alterations and reported the findings to attending physicians, who made the final clinical management decisions. RESULTS Between November 2016 and April 2019, 395 patients with 41 different tumors across 30 hospitals were included. Molecular analysis revealed actionable genetic alterations in 57 individuals (14.4%). Targeted therapies were advised for 23 and seven received a matched targeted therapy: two lung cancers (EML4-ALK and CD74-ROS1 fusion), three glioblastomas (EGFR point mutations), one oligodendroglioma (FGFR3-TACC3 fusion) and a prostate cancer (SND1-BRAF fusion). The outcomes included two tumor responses, one disease stabilization, one early withdrawal due to toxicity, one progression, and one unknown. CONCLUSION Despite the growing knowledge of cancer biology and its translation to drug development, the overall impact of personalized treatments remains low. Access to comprehensive molecular tests covering properly all known actionable alterations and programs for a wide access to targeted therapies seem to be critical steps.
Collapse
Affiliation(s)
- Paloma Navarro
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Carmen Beato
- Medical Oncology Department, Hospital Virgen de La Macarena, Seville, Spain
| | - Juan Francisco Rodriguez-Moreno
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Sergio Ruiz-Llorente
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- Departamento de Biomedicina y Biotecnología, Área de Genética, Universidad de Alcalá, Madrid, Spain
| | - Xabier Mielgo
- Medical Oncology Department , Hospital Universitario Fundacion Alcorcon, Madrid, Spain
| | - Estela Pineda
- Medical Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Miguel Navarro
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Gema Bruixola
- Medical Oncology Department, Hospital Clinico Universtario - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Tatiana P Grazioso
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain
- HM Hospitals Health Research Institute, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Antonio Viudez
- Medical Oncology Department, Hospital de Navarra, Pamplona, Spain
| | - Jose Fuster
- Medical Oncology Department, Hospital Universitario Son Espases, Palma, Spain
| | - Esther Nogueron
- Medical Oncology Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | | | - Carmen Balaña
- Instituto Catalán de Oncología-Badalona, Barcelona, Spain
| | | | | | | | | | | | - Jesus Garcia-Donas
- Laboratory of Innovation in Oncology; Gynecological, Genitourinary and Skin Tumor Unit, HM CIOCC (Clara Campal Comprehensive Cancer Centre), Sanchinarro HM Universitary Hospital, HM Hospitales, Madrid, Spain, Madrid, Spain.
- HM Faculty of Health Sciences, Camilo José Cela University, Madrid, Spain.
- HM Hospitals Health Research Institute, Madrid, Spain.
- Institute of Applied Molecular Medicine (IMMA), School of Medicine, Universidad San Pablo CEU, CEU Universities, Madrid, Spain.
| |
Collapse
|
12
|
Karaosmanoğlu O. Recurrent hepatocellular carcinoma is associated with the enrichment of MYC targets gene sets, elevated high confidence deleterious mutations and alternative splicing of DDB2 and BRCA1 transcripts. Adv Med Sci 2024; 70:17-26. [PMID: 39486583 DOI: 10.1016/j.advms.2024.10.004] [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: 01/15/2024] [Revised: 07/12/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
PURPOSE Recurrence is the main cause of hepatocellular carcinoma (HCC) related deaths. Underlying recurrence biology can be better understood by comparative analysis of the complete set of transcripts between recurrent and non-recurrent HCC. In this study, transcriptomic data (GSE56545) from 21 male patients diagnosed with either recurrent or non-recurrent HCC were reanalyzed to identify deregulated pathways, somatic mutations, fusion transcripts, alternative splicing events, and the immune context in recurrent HCC. MATERIALS AND METHODS DESeq2 was used for differential expression analysis, Mutect2 for somatic mutation analysis, Arriba and STAR-Fusion for fusion transcript analysis, and rMATs for alternative splicing analysis. RESULTS The results revealed that MYC targets gene sets (Hallmark_MYC_targets_V1 and Hallmark_MYC_targets_V2) were significantly enriched in recurrent HCC. Among the MYC targets, CBX3, NOP56, CDK4, NPM1, MCM5, MCM4 and PA2G4 upregulation was significantly associated with poor survival. Somatic mutation analysis demonstrated that the numbers of high confidence deleterious mutations were significantly increased in recurrent HCC. Alternative splicing-mediated production of non-functional DDB2 and oncogenic BRCA1 D11q were discovered in recurrent HCC. Finally, CD8+ T-cells were significantly decreased in recurrent HCC. CONCLUSIONS These results indicated that the enrichment of MYC targets gene sets is one of the most critical factors that leads to the development of recurrent HCC. In addition, elevated deleterious mutation numbers and alternative spliced DDB2 and BRCA1 isoforms have been identified as prominent contributors to increasing genomic instability in male patients with recurrent HCC.
Collapse
Affiliation(s)
- Oğuzhan Karaosmanoğlu
- Department of Biology, Kamil Özdağ Faculty of Science, Karamanoğlu Mehmetbey University, İbrahim Öktem Avenue, No. 124, 70200, Karaman, Turkey.
| |
Collapse
|
13
|
Meng F, Li H, Jin R, Yang A, Luo H, Li X, Wang P, Zhao Y, Chervova O, Tang K, Cheng S, Hu B, Li Y, Sheng J, Yang F, Carbone D, Chen K, Wang J. Spatial immunogenomic patterns associated with lymph node metastasis in lung adenocarcinoma. Exp Hematol Oncol 2024; 13:106. [PMID: 39468696 PMCID: PMC11514955 DOI: 10.1186/s40164-024-00574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) with lymph node (LN) metastasis is linked to poor prognosis, yet the underlying mechanisms remain largely undefined. This study aimed to elucidate the immunogenomic landscape associated with LN metastasis in LUAD. METHODS We employed broad-panel next-generation sequencing (NGS) on a cohort of 257 surgically treated LUAD patients to delineate the molecular landscape of primary tumors and identify actionable driver-gene alterations. Additionally, we used multiplex immunohistochemistry (mIHC) on a propensity score-matched cohort, which enabled us to profile the immune microenvironment of primary tumors in detail while preserving cellular metaclusters, interactions, and neighborhood functional units. By integrating data from NGS and mIHC, we successfully identified spatial immunogenomic patterns and developed a predictive model for LN metastasis, which was subsequently validated independently. RESULTS Our analysis revealed distinct immunogenomic alteration patterns associated with LN metastasis stages. Specifically, we observed increased mutation frequencies in genes such as PIK3CG and ATM in LN metastatic primary tumors. Moreover, LN positive primary tumors exhibited a higher presence of macrophage and regulatory T cell metaclusters, along with their enriched neighborhood units (p < 0.05), compared to LN negative tumors. Furthermore, we developed a novel predictive model for LN metastasis likelihood, designed to inform non-surgical treatment strategies, optimize personalized therapy plans, and potentially improve outcomes for patients who are ineligible for surgery. CONCLUSIONS This study offers a comprehensive analysis of the genetic and immune profiles in LUAD primary tumors with LN metastasis, identifying key immunogenomic patterns linked to metastatic progression. The predictive model derived from these insights marks a substantial advancement in personalized treatment, underscoring its potential to improve patient management.
Collapse
Affiliation(s)
- Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Ruoyi Jin
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Airong Yang
- Kanghui Biotechnology Co., Ltd, Shenyang, China
| | - Hao Luo
- Cancer Center, Daping Hospital Army Medical University, Chongqing, China
| | - Xiao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yaxing Zhao
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Olga Chervova
- University College London Cancer Institute, University College London, London, UK
| | - Kaicheng Tang
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Sida Cheng
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Jianpeng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - David Carbone
- James Thoracic Oncology Center, Ohio State University, Columbus, USA
| | - Kezhong Chen
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
| | - Jun Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
| |
Collapse
|
14
|
Lazar V, Raymond E, Magidi S, Bresson C, Wunder F, Berindan-Neagoe I, Tijeras-Rabaland A, Raynaud J, Onn A, Ducreux M, Batist G, Lassen U, Cilius Nielsen F, Schilsky RL, Rubin E, Kurzrock R. Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors. Ther Adv Med Oncol 2024; 16:17588359241289200. [PMID: 39429467 PMCID: PMC11487509 DOI: 10.1177/17588359241289200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Background Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options. Objective Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors. Design Exploring transcriptomic data (22,000 gene products) from the WINTHER trial (N = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available. Methods A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired t-test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes (K-mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot). Results We identified four key central hub genes-PLOD3, ARHGAP11A, RNF216, and CDCA8, for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts. Conclusion The expression of PLOD3, ARHGAP11A, RNF216, and CDCA8 constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions.
Collapse
Affiliation(s)
- Vladimir Lazar
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Eric Raymond
- Groupe Hospitalier Saint Joseph, Oncology Department Paris, France
| | - Shai Magidi
- Worldwide Innovative Network Association—WIN Consortium, 24, rue Albert Thuret, Chevilly-Larue 94850, France
| | - Catherine Bresson
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Fanny Wunder
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Ioana Berindan-Neagoe
- The Oncology Institute “Prof. Dr. Ion Chiricuta,” Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Jacques Raynaud
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Amir Onn
- Sheba Medical Center, Institute of Pulmonology, Tel HaShomer, Ramat-Gan, Israel
| | - Michel Ducreux
- Gustave Roussy, Department of Medical Oncology, Villejuif, France
- University Paris-Saclay, Department of Medical Oncology, Orsay, France
| | - Gerald Batist
- Segal Cancer Centre, Department of Oncology, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | | | | | | | - Eitan Rubin
- Ben-Gurion University of the Negev, The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Be’er-Sheva, Israel
| | | |
Collapse
|
15
|
Wahida A, Kurzrock R. The Molecular Tumor Board Turns 10: The Age of Complexity. Oncologist 2024:oyae271. [PMID: 39419766 DOI: 10.1093/oncolo/oyae271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Gene sequencing has brought a titanic of complex data into clinical precision oncology. Deciphering this complexity for practice requires new constructs. In 2014, the Molecular Tumor Board (MTB) was introduced into the literature by a publication in The Oncologist. Ten years later, MTBs have become globally established vehicles that integrate rapidly emerging "omic" information, helping to transform cancer management.
Collapse
Affiliation(s)
- Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Centre Munich, Neuherberg, Germany
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, United States
- WIN Consortium, Paris, France
| |
Collapse
|
16
|
Vasseur D, Bigot L, Beshiri K, Flórez-Arango J, Facchinetti F, Hollebecque A, Tselikas L, Aldea M, Blanc-Durand F, Gazzah A, Planchard D, Lacroix L, Pata-Merci N, Nobre C, Da Silva A, Nicotra C, Ngo-Camus M, Braye F, Nikolaev SI, Michiels S, Jules-Clement G, Olaussen KA, André F, Scoazec JY, Barlesi F, Ponce S, Soria JC, Besse B, Loriot Y, Friboulet L. Deciphering resistance mechanisms in cancer: final report of MATCH-R study with a focus on molecular drivers and PDX development. Mol Cancer 2024; 23:221. [PMID: 39363320 PMCID: PMC11451117 DOI: 10.1186/s12943-024-02134-4] [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: 07/08/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Understanding the resistance mechanisms of tumor is crucial for advancing cancer therapies. The prospective MATCH-R trial (NCT02517892), led by Gustave Roussy, aimed to characterize resistance mechanisms to cancer treatments through molecular analysis of fresh tumor biopsies. This report presents the genomic data analysis of the MATCH-R study conducted from 2015 to 2022 and focuses on targeted therapies. METHODS The study included resistant metastatic patients (pts) who accepted an image-guided tumor biopsy. After evaluation of tumor content (TC) in frozen tissue biopsies, targeted NGS (10 < TC < 30%) or Whole Exome Sequencing and RNA sequencing (TC > 30%) were performed before and/or after the anticancer therapy. Patient-derived xenografts (PDX) were established by implanting tumor fragments into NOD scid gamma mice and amplified up to five passages. RESULTS A total of 1,120 biopsies were collected from 857 pts with the most frequent tumor types being lung (38.8%), digestive (16.3%) and prostate (14.1%) cancer. Molecular targetable driver were identified in 30.9% (n = 265/857) of the patients, with EGFR (41.5%), FGFR2/3 (15.5%), ALK (11.7%), BRAF (6.8%), and KRAS (5.7%) being the most common altered genes. Furthermore, 66.0% (n = 175/265) had a biopsy at progression on targeted therapy. Among resistant cases, 41.1% (n = 72/175) had no identified molecular mechanism, 32.0% (n = 56/175) showed on-target resistance, and 25.1% (n = 44/175) exhibited a by-pass resistance mechanism. Molecular profiling of the 44 patients with by-pass resistance identified 51 variants, with KRAS (13.7%), PIK3CA (11.8%), PTEN (11.8%), NF2 (7.8%), AKT1 (5.9%), and NF1 (5.9%) being the most altered genes. Treatment was tailored for 45% of the patients with a resistance mechanism identified leading to an 11 months median extension of clinical benefit. A total of 341 biopsies were implanted in mice, successfully establishing 136 PDX models achieving a 39.9% success rate. PDX models are available for EGFR (n = 31), FGFR2/3 (n = 26), KRAS (n = 18), ALK (n = 16), BRAF (n = 6) and NTRK (n = 2) driven cancers. These models closely recapitulate the biology of the original tumors in term of molecular alterations and pharmacological status, and served as valuable models to validate overcoming treatment strategies. CONCLUSION The MATCH-R study highlights the feasibility of on purpose image guided tumor biopsies and PDX establishment to characterize resistance mechanisms and guide personalized therapies to improve outcomes in pre-treated metastatic patients.
Collapse
Affiliation(s)
- Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
- AMMICa UAR3655/US23, Gustave Roussy, Villejuif, France
| | - Ludovic Bigot
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Kristi Beshiri
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
| | | | | | - Antoine Hollebecque
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Department of Interventional Radiology, BIOTHERIS, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Mihaela Aldea
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | | | - Anas Gazzah
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
| | - David Planchard
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Ludovic Lacroix
- Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
- AMMICa UAR3655/US23, Gustave Roussy, Villejuif, France
| | | | - Catline Nobre
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Alice Da Silva
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Claudio Nicotra
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
| | - Maud Ngo-Camus
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
| | - Floriane Braye
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Sergey I Nikolaev
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Stefan Michiels
- Université Paris-Saclay, CESP, InsermVillejuif, France
- Office of Biostatistics and Epidemiology, Gustave Roussy, Villejuif, France
| | - Gérôme Jules-Clement
- Bioinformatics Core Facility, Gustave Roussy, Université Paris-Saclay, CNRS UMS 3655, Inserm US23, Villejuif, France
| | | | - Fabrice André
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Jean-Yves Scoazec
- Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
- AMMICa UAR3655/US23, Gustave Roussy, Villejuif, France
| | - Fabrice Barlesi
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Santiago Ponce
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Jean-Charles Soria
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Yohann Loriot
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France.
- Département d'Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France.
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France.
| | - Luc Friboulet
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France.
| |
Collapse
|
17
|
Wegmann R, Bankel L, Festl Y, Lau K, Lee S, Arnold F, Cappelletti V, Fehr A, Picotti P, Dedes KJ, Franzen D, Lenggenhager D, Bode PK, Zoche M, Moch H, Britschgi C, Snijder B. Molecular and functional landscape of malignant serous effusions for precision oncology. Nat Commun 2024; 15:8544. [PMID: 39358333 PMCID: PMC11447229 DOI: 10.1038/s41467-024-52694-8] [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: 06/14/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
Personalized treatment for patients with advanced solid tumors critically depends on the deep characterization of tumor cells from patient biopsies. Here, we comprehensively characterize a pan-cancer cohort of 150 malignant serous effusion (MSE) samples at the cellular, molecular, and functional level. We find that MSE-derived cancer cells retain the genomic and transcriptomic profiles of their corresponding primary tumors, validating their use as a patient-relevant model system for solid tumor biology. Integrative analyses reveal that baseline gene expression patterns relate to global ex vivo drug sensitivity, while high-throughput drug-induced transcriptional changes in MSE samples are indicative of drug mode of action and acquired treatment resistance. A case study exemplifies the added value of multi-modal MSE profiling for patients who lack genetically stratified treatment options. In summary, our study provides a functional multi-omics view on a pan-cancer solid tumor cohort and underlines the feasibility and utility of MSE-based precision oncology.
Collapse
Affiliation(s)
- Rebekka Wegmann
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lorenz Bankel
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
| | - Yasmin Festl
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Kate Lau
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Sohyon Lee
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Fabian Arnold
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Valentina Cappelletti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Aaron Fehr
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Konstantin J Dedes
- Department of Gynecology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Franzen
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Peter K Bode
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Martin Zoche
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Christian Britschgi
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
- Medical Oncology and Hematology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
- Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland.
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| |
Collapse
|
18
|
Kim TY, Kim SY, Kim JH, Jung HA, Choi YJ, Hwang IG, Cha Y, Lee GW, Lee YG, Kim TM, Lee SH, Lee S, Yun H, Choi YL, Yoon S, Han SW, Kim TY, Kim TW, Zang DY, Kang JH. Nationwide precision oncology pilot study: KOrean Precision Medicine Networking Group Study of MOlecular profiling-guided therapy based on genomic alterations in advanced solid tumors (KOSMOS) KCSG AL-20-05. ESMO Open 2024; 9:103709. [PMID: 39305545 PMCID: PMC11440300 DOI: 10.1016/j.esmoop.2024.103709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) has become widely available but molecular profiling-guided therapy (MGT) had not been well established in the real world due to lack of available therapies and expertise to match treatment. Our study was designed to test the feasibility of a nationwide platform of NGS-guided MGT recommended by a central molecular tumor board (cMTB) for metastatic solid tumors. PATIENTS AND METHODS Patients with advanced or metastatic solid tumors with available NGS results and without standard treatment were enrolled. The cMTB interpreted the patients' NGS reports and recommended the following: (i) investigational medicinal products (IMPs) approved in other indications; (ii) alternative treatments; (iii) clinical trials. The primary variables were the proportion of patients with actionable genomic alterations and those receiving MGT as per cMTB recommendations. Others included treatment duration (TD), overall response rate (ORR), disease control rate (DCR), and safety. RESULTS From February 2021 to February 2022, 193 cases [99 (51.3%) men; median age 58 years (range 24-88 years); median line of previous treatment 3 (range 0-9)] from 29 sites were enrolled for 60 cMTB sessions. The median time from case submission to cMTB discussion was 7 days (range 2-20 days), and to IMP treatment initiation was 28 days (range 14-90 days). Actionable genetic alterations were found in 145 patients (75.1%). A total of 89 (46.1%) patients received actual dosing of IMPs, and 10 (5.2%) were enrolled in cMTB-recommended clinical trials, achieving an MGT rate of 51.3%. ORR and DCR of IMPs were 10.1% and 72.5%, respectively. The median TD was 3.5 months [95% confidence interval (CI) 2.8-5.5 months], and the 4-month TD rate was 44.9%. The median overall survival of patients who received IMPs was 6.9 months (95% CI 5.2-10.0 months). CONCLUSION KOSMOS confirmed the feasibility of MGT recommended by the cMTB, achieving a high MGT match rate and promising effectiveness in heavily pretreated advanced cancer patients.
Collapse
Affiliation(s)
- T-Y Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - S Y Kim
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - J H Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam.
| | - H A Jung
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul
| | - Y J Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul
| | - I G Hwang
- Department of Internal Medicine, Chung-Ang University College of Medicine, Chung-Ang University Hospital, Seoul
| | - Y Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang
| | - G-W Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju
| | - Y-G Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Seoul
| | - T M Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - S-H Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul
| | - S Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul
| | - H Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul
| | - Y L Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - S Yoon
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - S W Han
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - T-Y Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - T W Kim
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - D Y Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical Center, Anyang
| | - J H Kang
- Department of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
van Schaik LF, Engelhardt EG, Wilthagen EA, Steeghs N, Fernández Coves A, Joore MA, van Harten WH, Retèl VP. Factors for a broad technology assessment of comprehensive genomic profiling in advanced cancer, a systematic review. Crit Rev Oncol Hematol 2024; 202:104441. [PMID: 39002790 DOI: 10.1016/j.critrevonc.2024.104441] [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: 04/15/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024] Open
Abstract
Comprehensive Genomic Profiling (CGP) allows for the identification of many targets. Reimbursement decision-making is, however, challenging because besides the health benefits of on-label treatments and costs, other factors related to diagnostic and treatment pathways may also play a role. The aim of this study was to identify which other factors are relevant for the technology assessment of CGP and to summarize the available evidence for these factors. After a scoping search and two expert sessions, five factors were identified: feasibility, test journey, wider implications of diagnostic results, organisation of laboratories, and "scientific spillover". Subsequently, a systematic search identified 83 studies collecting mainly evidence for the factors "test journey" and "wider implications of diagnostic results". Its nature was, however, of limited value for decision-making. We recommend the use of comparative strategies, uniformity in outcome definitions, and the inclusion of a comprehensive set of factors in future evidence generation.
Collapse
Affiliation(s)
- L F van Schaik
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| | - E G Engelhardt
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands.
| | - E A Wilthagen
- Scientific Information Service, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - N Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - A Fernández Coves
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - M A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - W H van Harten
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands.
| | - V P Retèl
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
20
|
Rassy E, Mosele MF, Di Meglio A, Pistilli B, Andre F. Precision oncology in patients with breast cancer: towards a 'screen and characterize' approach. ESMO Open 2024; 9:103716. [PMID: 39303452 PMCID: PMC11439525 DOI: 10.1016/j.esmoop.2024.103716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- E Rassy
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Oncostat U1018, Inserm, Université Paris-Saclay, Equipe labellisée Ligue Contre le Cancer, Villejuif
| | - M F Mosele
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - A Di Meglio
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - B Pistilli
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; INSERM U1279, Gustave Roussy, Villejuif, France
| | - F Andre
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif.
| |
Collapse
|
21
|
Reina C, Šabanović B, Lazzari C, Gregorc V, Heeschen C. Unlocking the future of cancer diagnosis - promises and challenges of ctDNA-based liquid biopsies in non-small cell lung cancer. Transl Res 2024; 272:41-53. [PMID: 38838851 DOI: 10.1016/j.trsl.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
The advent of liquid biopsies has brought significant changes to the diagnosis and monitoring of non-small cell lung cancer (NSCLC), presenting both promise and challenges. Molecularly targeted drugs, capable of enhancing survival rates, are now available to around a quarter of NSCLC patients. However, to ensure their effectiveness, precision diagnosis is essential. Circulating tumor DNA (ctDNA) analysis as the most advanced liquid biopsy modality to date offers a non-invasive method for tracking genomic changes in NSCLC. The potential of ctDNA is particularly rooted in its ability to furnish comprehensive (epi-)genetic insights into the tumor, thereby aiding personalized treatment strategies. One of the key advantages of ctDNA-based liquid biopsies in NSCLC is their ability to capture tumor heterogeneity. This capability ensures a more precise depiction of the tumor's (epi-)genomic landscape compared to conventional tissue biopsies. Consequently, it facilitates the identification of (epi-)genetic alterations, enabling informed treatment decisions, disease progression monitoring, and early detection of resistance-causing mutations for timely therapeutic interventions. Here we review the current state-of-the-art in ctDNA-based liquid biopsy technologies for NSCLC, exploring their potential to revolutionize clinical practice. Key advancements in ctDNA detection methods, including PCR-based assays, next-generation sequencing (NGS), and digital PCR (dPCR), are discussed, along with their respective strengths and limitations. Additionally, the clinical utility of ctDNA analysis in guiding treatment decisions, monitoring treatment response, detecting minimal residual disease, and identifying emerging resistance mechanisms is examined. Liquid biopsy analysis bears the potential of transforming NSCLC management by enabling non-invasive monitoring of Minimal Residual Disease and providing early indicators for response to targeted treatments including immunotherapy. Furthermore, considerations regarding sample collection, processing, and data interpretation are highlighted as crucial factors influencing the reliability and reproducibility of ctDNA-based assays. Addressing these challenges will be essential for the widespread adoption of ctDNA-based liquid biopsies in routine clinical practice, ultimately paving the way toward personalized medicine and improved outcomes for patients with NSCLC.
Collapse
Affiliation(s)
- Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Chiara Lazzari
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Vanesa Gregorc
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Christopher Heeschen
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy;.
| |
Collapse
|
22
|
De Landtsheer S, Badkas A, Kulms D, Sauter T. Model ensembling as a tool to form interpretable multi-omic predictors of cancer pharmacosensitivity. Brief Bioinform 2024; 25:bbae567. [PMID: 39494610 PMCID: PMC11532660 DOI: 10.1093/bib/bbae567] [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: 07/10/2024] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Stratification of patients diagnosed with cancer has become a major goal in personalized oncology. One important aspect is the accurate prediction of the response to various drugs. It is expected that the molecular characteristics of the cancer cells contain enough information to retrieve specific signatures, allowing for accurate predictions based solely on these multi-omic data. Ideally, these predictions should be explainable to clinicians, in order to be integrated in the patients care. We propose a machine-learning framework based on ensemble learning to integrate multi-omic data and predict sensitivity to an array of commonly used and experimental compounds, including chemotoxic compounds and targeted kinase inhibitors. We trained a set of classifiers on the different parts of our dataset to produce omic-specific signatures, then trained a random forest classifier on these signatures to predict drug responsiveness. We used the Cancer Cell Line Encyclopedia dataset, comprising multi-omic and drug sensitivity measurements for hundreds of cell lines, to build the predictive models, and validated the results using nested cross-validation. Our results show good performance for several compounds (Area under the Receiver-Operating Curve >79%) across the most frequent cancer types. Furthermore, the simplicity of our approach allows to examine which omic layers have a greater importance in the models and identify new putative markers of drug responsiveness. We propose several models based on small subsets of transcriptional markers with the potential to become useful tools in personalized oncology, paving the way for clinicians to use the molecular characteristics of the tumors to predict sensitivity to therapeutic compounds.
Collapse
Affiliation(s)
- Sébastien De Landtsheer
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Apurva Badkas
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technische Universität-Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases, Technische Universität-Dresden, 01307 Dresden, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
23
|
Hung LJ, Huang CY, Tung KC, Chen JS, Huang WK, Hsu CC, Fang YF, Wang CL, Liu PC, Yeh KY, Chang PH, Chang JWC, Lin YC, Huang SF, Chou WC. Comprehensive genomic profiling in multiple cancer types: A comparative analysis of the National Biobank Consortium of Taiwan and clinical practice cohorts. J Formos Med Assoc 2024:S0929-6646(24)00405-4. [PMID: 39244401 DOI: 10.1016/j.jfma.2024.09.001] [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: 05/08/2024] [Revised: 07/30/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND This retrospective study analyzed tumor tissue profiling data to assess the potential of comprehensive genomic profiling (CGP) for patient care across diverse solid tumors. MATERIAL AND METHODS Patients with newly diagnosed or recurrent stage IIIB or IV lung adenocarcinoma with a null immunophenotype and esophageal, gastric, pancreatic, or bile duct cancer between January 2020 and July 2023 at two medical centers in Taiwan were included. One cohort was a part of the National Biobank Consortium of Taiwan project, whereas the other consisted of patients undergoing routine clinical practice. Tumor samples were subjected to CGP using FoundationOne®CDx, with therapeutic implications determined using OncoKB classification. RESULTS FoundationOne®CDx testing of 574 patients was successful in 456 (79.4%) patients. Clinically actionable genomic alterations were detected in 21.1% (96/456) of the patients, including 17.5%, 2.9%, and 0.7% of patients with evidence levels 1, 2, and 3, respectively. Lung adenocarcinoma accounted for the largest proportion of samples with at least one actionable gene alteration (63.2%), followed by bile duct (26.9%), gastric (17.6%), esophageal (4.0%), and pancreatic (3.1%) cancers. Based on CGP results, 43 patients (9.4%) received matched targeted therapy. The median overall survival of patients who received matched therapy or not was 26.1 months (95% confidence interval (CI), 16.7-35.5 months) and 10.6 months (95% CI, 8.1-13.1 months; hazard ratio, 0.28, 95% CI, 0.14-0.55, p < 0.001), respectively. CONCLUSIONS This study provides comprehensive insights into the genomic profiles of diverse cancers in Taiwan, highlighting the crucial role of CGP in identifying actionable genomic alterations and guiding effective therapeutic strategies in real-world practice.
Collapse
Affiliation(s)
- Ling-Jen Hung
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Kai-Che Tung
- Patient Journey Solution Division, Roche Products Ltd., Taipei, Taiwan; Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jen-Shi Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Chung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yueh-Fu Fang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Liang Wang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Ping-Chi Liu
- Division of Thoracic Oncology, Department of Thoracic Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Kun-Yun Yeh
- Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Pei-Hung Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
24
|
Hernando-Calvo A, Han M, Ayodele O, Wang BX, Bruce JP, Abbas-Aghababazadeh F, Vila-Casadesús M, Sanz-Garcia E, Yang SYC, Berman HK, Vivancos A, Lam B, Lungu I, Salawu A, Stayner LA, Haibe-Kains B, Bedard PL, Avery L, Razak ARA, Pugh TJ, Spreafico A, Siu LL, Hansen AR. A Phase II, Open-Label, Randomized Trial of Durvalumab With Olaparib or Cediranib in Patients With Mismatch Repair-Proficient Colorectal or Pancreatic Cancer. Clin Colorectal Cancer 2024; 23:272-284.e9. [PMID: 38960798 DOI: 10.1016/j.clcc.2024.05.002] [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: 02/28/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The use of immunotherapy in mismatch repair proficient colorectal cancer (pMMR-CRC) or pancreatic adenocarcinoma (PDAC) is associated with limited efficacy. DAPPER (NCT03851614) is a phase 2, basket study randomizing patients with pMMR CRC or PDAC to durvalumab with olaparib (durvalumab + olaparib) or durvalumab with cediranib (durvalumab + cediranib). METHODS PDAC or pMMR-CRC patients were randomized to either durvalumab+olaparib (arm A), or durvalumab + cediranib (arm B). Co-primary endpoints included pharmacodynamic immune changes in the tumor microenvironment (TME) and safety. Objective response rate, progression-free survival (PFS) and overall survival (OS) were determined. Paired tumor samples were analyzed by multiplexed immunohistochemistry and RNA-sequencing. RESULTS A total of 31 metastatic pMMR-CRC patients were randomized to arm A (n = 16) or B (n = 15). In 28 evaluable patients, 3 patients had stable disease (SD) (2 patients treated with durvalumab + olaparib and 1 patient treated with durvalumab + cediranib) while 25 had progressive disease (PD). Among patients with PDAC (n = 19), 9 patients were randomized to arm A and 10 patients were randomized to arm B. In 18 evaluable patients, 1 patient had a partial response (unconfirmed) with durvalumab + cediranib, 1 patient had SD with durvalumab + olaparib while 16 had PD. Safety profile was manageable and no grade 4-5 treatment-related adverse events were observed in either arm A or B. No significant changes were observed for CD3+/CD8+ immune infiltration in on-treatment biopsies as compared to baseline for pMMR-CRC and PDAC independent of treatment arms. Increased tumor-infiltrating lymphocytes at baseline, low baseline CD68+ cells and different immune gene expression signatures at baseline were associated with outcomes. CONCLUSIONS In patients with pMMR-CRC or PDAC, durvalumab + olaparib and durvalumab + cediranib showed limited antitumor activity. Different immune components of the TME were associated with treatment outcomes.
Collapse
Affiliation(s)
- Alberto Hernando-Calvo
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ming Han
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Olubukola Ayodele
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ben X Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey P Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | - Enrique Sanz-Garcia
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - S Y Cindy Yang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Hal K Berman
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ana Vivancos
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Bernard Lam
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Ilinca Lungu
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Abdulazeez Salawu
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Lee-Anne Stayner
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Department of Computer Science, University of Toronto, Toronto, Ontario, Canada; Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - Philippe L Bedard
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Lisa Avery
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Albiruni R A Razak
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Lillian L Siu
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Aaron R Hansen
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Zhang J, Xu S. High aggressiveness of papillary thyroid cancer: from clinical evidence to regulatory cellular networks. Cell Death Discov 2024; 10:378. [PMID: 39187514 PMCID: PMC11347646 DOI: 10.1038/s41420-024-02157-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
The global incidence of thyroid cancer has increased over recent decades. Papillary thyroid cancer (PTC) is the most common type of thyroid cancer and accounts for nearly 90% of all cases. Typically, PTC has a good prognosis. However, some PTC variants exhibit more aggressive behaviour, which significantly increases the risk of postoperative recurrence. Over the past decade, the high metastatic potential of PTC has drawn the attention of many researchers and these studies have provided useful molecular markers for improved diagnosis, risk stratification and clinical approaches. The aim of this review is to discuss the progress in epidemiology, metastatic features, risk factors and molecular mechanisms associated with PTC aggressiveness. We present a detailed picture showing that epithelial-to-mesenchymal transition, cancer metabolic reprogramming, alterations in important signalling pathways, epigenetic aberrations and the tumour microenvironment are crucial drivers of PTC metastasis. Further research is needed to more fully elucidate the pathogenesis and biological behaviour underlying the aggressiveness of PTC.
Collapse
Affiliation(s)
- Junsi Zhang
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou, China.
| |
Collapse
|
26
|
Mechahougui H, Gutmans J, Colarusso G, Gouasmi R, Friedlaender A. Advances in Personalized Oncology. Cancers (Basel) 2024; 16:2862. [PMID: 39199633 PMCID: PMC11352922 DOI: 10.3390/cancers16162862] [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: 06/16/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Advances in next-generation sequencing (NGS) have catalyzed a paradigm shift in cancer treatment, steering the focus from conventional, organ-specific protocols to precision medicine. Emerging targeted therapies offer a cutting-edge approach to cancer treatment, while companion diagnostics play an essential role in aligning therapeutic choices with specific molecular changes identified through NGS. Despite these advances, interpreting the clinical implications of a rapidly expanding catalog of genetic mutations remains a challenge. The selection of therapies in the presence of multiple mutations requires careful clinical judgment, supported by quality-centric genomic testing that emphasizes actionable mutations. Molecular tumor boards can play an increasing role in assimilating genomic data into clinical trials, thereby refining personalized treatment approaches and improving patient outcomes.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - Gina Colarusso
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France
| | | |
Collapse
|
27
|
Louie BH, Kato S, Lim JS, Kim KH, Lim HJ, Okamura R, Lee S, Kim L, Sicklick JK, Lippman SM, Kurzrock R. Molecular Tumor Board for Unicorns: Outcomes for rare and ultra-rare cancers using an N-of-One personalized treatment strategy. iScience 2024; 27:110465. [PMID: 39148716 PMCID: PMC11324991 DOI: 10.1016/j.isci.2024.110465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 05/06/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Treatment of rare/ultra-rare tumors is an unmet need due to a lack of standardized therapies and clinical trials. We developed the Molecular Tumor Board (MTB), a multidisciplinary team that integrates molecular profiling to generate personalized, N-of-One treatments for advanced cancers. This study evaluates 112 patients with rare/ultra-rare tumors who presented to the MTB and were evaluable for clinical therapeutic outcome. Overall, 46/112 patients (41%) received a treatment regimen with a high degree of matching between tumor molecular alterations and drugs given (reflected by a high Matching Score (≥50%)). Patients with a high versus low Matching Score experienced significantly longer progression-free survival (p = 0.005) and overall survival (p = 0.047), and higher rates of clinical benefit (stable disease ≥6 months, partial response, or complete response) (54% vs. 32% p = 0.027). The MTB facilitated personalized N-of-One matching of drugs to tumor molecular alterations, which was associated with improved clinical outcomes in patients with rare/ultra-rare cancers.
Collapse
Affiliation(s)
- Bryan H Louie
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jordan S Lim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Ki Hwan Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Hyo Jeong Lim
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Ryosuke Okamura
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Lisa Kim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, and Department of Pharmacology, UC San Diego Health Sciences, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, Paris, France
- Medical College of Wisconsin Cancer Center and Genomic Sciences and Precision Medicine Center, Milwaukee, WI, USA
- University of Nebraska, Omaha, NE, USA
| |
Collapse
|
28
|
Kuehn JC, Metzger P, Neidert N, Matysiak U, Gräßel L, Philipp U, Bleul S, Pauli T, Falkenstein J, Bertemes H, Cysar S, Hess ME, Frey AV, Duque-Afonso J, Schorb E, Machein M, Beck J, Schnell O, von Bubnoff N, Illert AL, Peters C, Brummer T, Prinz M, Miething C, Becker H, Lassmann S, Werner M, Börries M, Duyster J, Heiland DH, Sankowski R, Scherer F. Comprehensive genetic profiling and molecularly guided treatment for patients with primary CNS tumors. NPJ Precis Oncol 2024; 8:180. [PMID: 39143272 PMCID: PMC11324882 DOI: 10.1038/s41698-024-00674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
Despite major advances in molecular profiling and classification of primary brain tumors, personalized treatment remains limited for most patients. Here, we explored the feasibility of individual molecular profiling and the efficacy of biomarker-guided therapy for adult patients with primary brain cancers in the real-world setting within the molecular tumor board Freiburg, Germany. We analyzed genetic profiles, personalized treatment recommendations, and clinical outcomes of 102 patients with 21 brain tumor types. Alterations in the cell cycle, BRAF, and mTOR pathways most frequently led to personalized treatment recommendations. Molecularly informed therapies were recommended in 71% and implemented in 32% of patients with completed molecular diagnostics. The disease control rate following targeted treatment was 50% and the overall response rate was 30%, with a progression-free survival 2/1 ratio of at least 1.3 in 31% of patients. This study highlights the efficacy of molecularly guided treatment and the need for biomarker-stratified trials in brain cancers.
Collapse
Affiliation(s)
- Julia C Kuehn
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicolas Neidert
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Uta Matysiak
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Linda Gräßel
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrike Philipp
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Bleul
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Pauli
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Falkenstein
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Henriette Bertemes
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stepan Cysar
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Elena Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Anna Verena Frey
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jesús Duque-Afonso
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elisabeth Schorb
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcia Machein
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Anna L Illert
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Department of Medicine III, Faculty of Medicine, Klinikum Rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Christoph Peters
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tilman Brummer
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Becker
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Silke Lassmann
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Börries
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roman Sankowski
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Scherer
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany.
| |
Collapse
|
29
|
Hayashi N, Ono M, Fukada I, Yamazaki M, Sato N, Hosonaga M, Wang X, Kaneko K, Arakawa H, Habano E, Kuga A, Kataoka A, Ueki A, Kiyotani K, Tonooka A, Takeuchi K, Kogawa T, Kitano S, Takano T, Watanabe M, Mori S, Takahashi S. Addressing the knowledge gap in the genomic landscape and tailored therapeutic approaches to adolescent and young adult cancers. ESMO Open 2024; 9:103659. [PMID: 39137480 PMCID: PMC11369407 DOI: 10.1016/j.esmoop.2024.103659] [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: 04/01/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Adolescents and young adults (AYAs) represent a small proportion of patients with cancer. The genomic profiles of AYA patients with cancer are not well-studied, and outcomes of genome-matched therapies remain largely unknown. PATIENTS AND METHODS We investigated differences between Japanese AYA and older adult (OA) patients in genomic alterations, therapeutic evidence levels, and genome-matched therapy usage by cancer type. We also assessed treatment outcomes. RESULTS AYA patients accounted for 8.3% of 876 cases. Microsatellite instability-high and/or tumor mutation burden was less common in AYA patients (1.4% versus 7.7% in OA; P = 0.05). However, BRCA1 alterations were more common in AYA patients with breast cancer (27.3% versus 1.7% in OA; P = 0.01), as were MYC alterations in AYA patients with colorectal cancer (23.5% versus 5.8% in OA; P = 0.02) and sarcoma (31.3% versus 3.4% in OA; P = 0.01). Genome-matched therapy use was similar between groups, with overall survival tending to improve in both. However, in AYA patients, the small number of patients prevented statistical significance. Comprehensive genomic profiling-guided genome-matched therapy yielded encouraging results, with progression-free survival of 9.0 months in AYA versus 3.7 months in OA patients (P = 0.59). CONCLUSION Our study suggests that tailored therapeutic approaches can benefit cancer patients regardless of age.
Collapse
Affiliation(s)
- N Hayashi
- Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - M Ono
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo.
| | - I Fukada
- Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - M Yamazaki
- Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - N Sato
- Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - M Hosonaga
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Tokyo
| | - X Wang
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - K Kaneko
- Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - H Arakawa
- Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - E Habano
- Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - A Kuga
- Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - A Kataoka
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Tokyo
| | - A Ueki
- Department of Clinical Genetic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - K Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Laboratory of Immunogenomics, The Center for Intractable Diseases and ImmunoGenomics (CiDIG), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka
| | - A Tonooka
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Pathology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - K Takeuchi
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Pathology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - T Kogawa
- Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - S Kitano
- Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - T Takano
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Tokyo
| | - M Watanabe
- Total Care Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| | - S Mori
- Project for Development of Innovative Research on Cancer Therapeutics, The Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - S Takahashi
- Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo; Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-ku, Tokyo
| |
Collapse
|
30
|
Sobsey CA, Froehlich BC, Mitsa G, Ibrahim S, Popp R, Zahedi RP, de Bruin EC, Borchers CH, Batist G. mTORC1-Driven Protein Translation Correlates with Clinical Benefit of Capivasertib within a Genetically Preselected Cohort of PIK3CA-Altered Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:2058-2074. [PMID: 38954770 PMCID: PMC11320025 DOI: 10.1158/2767-9764.crc-24-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/12/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Capivasertib is a potent selective inhibitor of AKT. It was recently FDA approved in combination with fulvestrant to treat HR+, HER2-negative breast cancers with certain genetic alteration(s) activating the PI3K pathway. In phase I trials, heavily pretreated patients with tumors selected for activating PI3K pathway mutations treated with capivasertib monotherapy demonstrated objective response rates of <30%. We investigated the proteomic profile associated with capivasertib response in genetically preselected patients and cancer cell lines. We analyzed samples from 16 PIK3CA-mutated patient tumors collected prior to capivasertib monotherapy in the phase I trial. PI3K pathway proteins were precisely quantified with immuno-Matrix-Assisted Laser Desorption/Ionization-mass spectrometry (iMALDI-MS). Global proteomic profiles were also obtained. Patients were classified according to response to capivasertib monotherapy: "clinical benefit (CB)" (≥12 weeks without progression, n = 7) or "no clinical benefit (NCB)" (progression in <12 weeks, n = 9). Proteins that differed between the patient groups were subsequently quantified in AKT1- or PIK3CA-altered breast cancer cell lines with varying capivasertib sensitivity. The measured concentrations of AKT1 and AKT2 varied among the PIK3CA-mutated tumors but did not differ between the CB and NCB groups. However, analysis of the global proteome data showed that translational activity was higher in tumors of the NCB vs. CB group. When reproducibly quantified by validated LC-MRM-MS assays, the same proteins of interest similarly distinguished between capivasertib-sensitive versus -resistant cell lines. The results provide further evidence that increased mTORC1-driven translation functions as a mechanism of resistance to capivasertib monotherapy. Protein concentrations may offer additional insights for patient selection for capivasertib, even among genetically preselected patients. SIGNIFICANCE Capivasertib's first-in-class FDA approval demonstrates its promise, yet there remains an opportunity to optimize its use. Our results provide new evidence that proteomics can stratify genetically preselected patients on clinical benefit. Characterization of the same profile in cell lines furnishes additional validation. Among PIK3CA-altered tumors, increased mTORC1-driven translation appears to confer intrinsic resistance. Assessing mTORC1 activation could therefore prove a useful complement to the existing genetic selection strategy for capivasertib.
Collapse
Affiliation(s)
- Constance A. Sobsey
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| | - Bjoern C. Froehlich
- University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria, Victoria, BC, Canada.
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
| | - Georgia Mitsa
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| | - Sahar Ibrahim
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| | | | - Rene P. Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, Canada.
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.
- CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.
| | | | - Christoph H. Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
| | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- McGill Centre for Translational Research in Cancer, Lady Davis Institute, Montreal, QC, Canada.
| |
Collapse
|
31
|
Pastò B, Buzzatti G, Schettino C, Malapelle U, Bergamini A, De Angelis C, Musacchio L, Dieci MV, Kuhn E, Lambertini M, Passarelli A, Toss A, Farolfi A, Roncato R, Capoluongo E, Vida R, Pignata S, Callari M, Baldassarre G, Bartoletti M, Gerratana L, Puglisi F. Unlocking the potential of Molecular Tumor Boards: from cutting-edge data interpretation to innovative clinical pathways. Crit Rev Oncol Hematol 2024; 199:104379. [PMID: 38718940 DOI: 10.1016/j.critrevonc.2024.104379] [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: 01/30/2024] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
The emerging era of precision medicine is characterized by an increasing availability of targeted anticancer therapies and by the parallel development of techniques to obtain more refined molecular data, whose interpretation may not always be straightforward. Molecular tumor boards gather various professional figures, in order to leverage the analysis of molecular data and provide prognostic and predictive insights for clinicians. In addition to healthcare development, they could also become a tool to promote knowledge and research spreading. A growing body of evidence on the application of molecular tumor boards to clinical practice is forming and positive signals are emerging, although a certain degree of heterogeneity exists. This work analyzes molecular tumor boards' potential workflows, figures involved, data sources, sample matrices and eligible patients, as well as available evidence and learning examples. The emerging concept of multi-institutional, disease-specific molecular tumor boards is also considered by presenting two ongoing nationwide experiences.
Collapse
Affiliation(s)
- Brenno Pastò
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giulia Buzzatti
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy
| | - Clorinda Schettino
- Clinical Trials Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli 80131, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Napoli 80131, Italy
| | - Alice Bergamini
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milano 20132, Italy; Unit of Obstetrics and Gynaecology, IRCCS San Raffaele Scientific Institute, Milano 20132, Italy
| | - Carmine De Angelis
- Oncology Unit - Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli 80131, Italy
| | - Lucia Musacchio
- Department of Women and Child Health, Division of Gynaecologic Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Roma 00168, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova 35122, Italy; Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova 35128, Italy
| | - Elisabetta Kuhn
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano 20122, Italy; Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova 16132, Italy
| | - Anna Passarelli
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | - Angela Toss
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena 41124, Italy; Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena 41124, Italy
| | - Alberto Farolfi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola 47014, Italy
| | - Rossana Roncato
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Ettore Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Napoli 80131, Italy; Clinical Pathology Unit, Azienda Ospedaliera San Giovanni Addolorata, Roma 00184, Italy
| | - Riccardo Vida
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Sandro Pignata
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | | | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Michele Bartoletti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Lorenzo Gerratana
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy.
| | - Fabio Puglisi
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| |
Collapse
|
32
|
Ren T, Zheng Y, Liu F, Liu C, Zhang B, Ren H, Gao X, Wei Y, Sun Q, Huang H. Identification and Validation of JAM-A as a Novel Prognostic and Immune Factor in Human Tumors. Biomedicines 2024; 12:1423. [PMID: 39061997 PMCID: PMC11275048 DOI: 10.3390/biomedicines12071423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Junctional adhesion molecule-A (JAM-A), also known as F11 receptor (F11R), is a transmembrane glycoprotein that is involved in various biological processes, including cancer initiation and progression. However, the functional characteristics and significance of JAM-A in pan-cancer remain unexplored. In this study, we used multiple databases to gain a comprehensive understanding of JAM-A in human cancers. JAM-A was widely expressed in various tissues, mainly located on the microtubules and cell junctions. Aberrant expression of JAM-A was detected in multiple cancers at both mRNA and protein levels, which can be correlated with poorer prognosis and may be attributed to genetic alterations and down-regulated DNA methylation. JAM-A expression was also associated with immune infiltration and may affect immunotherapy responses in several cancers. Functional enrichment analysis indicated that JAM-A participated in tight junction and cancer-related pathways. In vitro experiments verified that JAM-A knockdown suppressed the proliferation and migration abilities of breast cancer cells and liver cancer cells. Overall, our study suggests that JAM-A is a pan-cancer regulator and a potential biomarker for predicting prognosis and immune-therapeutic responses for different tumors.
Collapse
Affiliation(s)
- Tianyi Ren
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China; (T.R.); (C.L.); (H.R.)
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - You Zheng
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - Feichang Liu
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Chenyu Liu
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China; (T.R.); (C.L.); (H.R.)
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - Bo Zhang
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - He Ren
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China; (T.R.); (C.L.); (H.R.)
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - Xinyue Gao
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - Yuexian Wei
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, China
| | - Qiang Sun
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Beijing 100071, China; (Y.Z.); (F.L.); (B.Z.); (X.G.); (Y.W.)
| | - Hongyan Huang
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China; (T.R.); (C.L.); (H.R.)
| |
Collapse
|
33
|
El-Sayed MM, Bianco JR, Li Y, Fabian Z. Tumor-Agnostic Therapy-The Final Step Forward in the Cure for Human Neoplasms? Cells 2024; 13:1071. [PMID: 38920700 PMCID: PMC11201516 DOI: 10.3390/cells13121071] [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: 05/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.
Collapse
Affiliation(s)
| | | | | | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (M.M.E.-S.); (J.R.B.); (Y.L.)
| |
Collapse
|
34
|
Dhruba SR, Sahni S, Wang B, Wu D, Rajagopal PS, Schmidt Y, Shulman ED, Sinha S, Sammut SJ, Caldas C, Wang K, Ruppin E. The expression patterns of different cell types and their interactions in the tumor microenvironment are predictive of breast cancer patient response to neoadjuvant chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598770. [PMID: 39372749 PMCID: PMC11451622 DOI: 10.1101/2024.06.14.598770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The tumor microenvironment (TME) is a complex ecosystem of diverse cell types whose interactions govern tumor growth and clinical outcome. While the TME's impact on immunotherapy has been extensively studied, its role in chemotherapy response remains less explored. To address this, we developed DECODEM (DEcoupling Cell-type-specific Outcomes using DEconvolution and Machine learning), a generic computational framework leveraging cellular deconvolution of bulk transcriptomics to associate the gene expression of individual cell types in the TME with clinical response. Employing DECODEM to analyze the gene expression of breast cancer (BC) patients treated with neoadjuvant chemotherapy, we find that the gene expression of specific immune cells (myeloid, plasmablasts, B-cells) and stromal cells (endothelial, normal epithelial, CAFs) are highly predictive of chemotherapy response, going beyond that of the malignant cells. These findings are further tested and validated in a single-cell cohort of triple negative breast cancer. To investigate the possible role of immune cell-cell interactions (CCIs) in mediating chemotherapy response, we extended DECODEM to DECODEMi to identify such CCIs, validated in single-cell data. Our findings highlight the importance of active pre-treatment immune infiltration for chemotherapy success. The tools developed here are made publicly available and are applicable for studying the role of the TME in mediating response from readily available bulk tumor expression in a wide range of cancer treatments and indications.
Collapse
Affiliation(s)
- Saugato Rahman Dhruba
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sahil Sahni
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Binbin Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Di Wu
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Padma Sheila Rajagopal
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yael Schmidt
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eldad D. Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Carlos Caldas
- Institute of Metabolic Science, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Kun Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Takamizawa S, Koyama T, Sunami K, Sudo K, Hirata M, Kubo T, Tao K, Cho H, Narita Y, Kato K, Yamazaki N, Ohe Y, Okusaka T, Matsui Y, Ogawa C, Yonemori K, Yamamoto N. Identification of barriers to implementation of precision oncology in patients with rare cancers. Cancer Sci 2024; 115:2023-2035. [PMID: 38538548 PMCID: PMC11145155 DOI: 10.1111/cas.16165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 06/04/2024] Open
Abstract
Established treatment options for rare cancers are limited by the small number of patients. The current comprehensive genomic profiling (CGP) testing might not fully exploit opportunities for precision oncology in patients with rare cancers. Therefore, we aimed to explore the factors associated with CGP testing utility in rare cancers and identify barriers to implementing precision oncology. Patients who underwent CGP testing at our institution between September 2019 and June 2021 were enrolled in this retrospective study. Based on their results, the patients received molecularly targeted drugs or immune checkpoint inhibitors. Univariate and multivariate analyses evaluated the association between patient characteristics and the proportion of patients receiving molecularly targeted drugs. Overall, 790 patients underwent CGP testing. Among them, 333 patients with rare cancers were identified, of whom 278 (83.5%) had actionable genomic alterations, 127 (38.1%) had druggable genomic alterations, and 25 (7.5%) received genomically matched therapy. The proportion of patients receiving molecularly targeted drugs was significantly higher among those with treatment options with evidence levels A-D (8.7%) than those without treatment options with evidence levels A-D (2.9%). A potential barrier to CGP testing utility in rare cancers is the limited number of molecularly targeted drugs with clinical evidence. We propose that CGP testing be performed in patients with rare cancers who have treatment options with evidence levels A-D to maximize CGP testing utility in real-world practice.
Collapse
Affiliation(s)
- Shigemasa Takamizawa
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Takafumi Koyama
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Kuniko Sunami
- Department of Laboratory MedicineNational Cancer Center HospitalTokyoJapan
| | - Kazuki Sudo
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Makoto Hirata
- Department of Genetic Medicine and ServicesNational Cancer Center HospitalTokyoJapan
| | - Takashi Kubo
- Division of Translational GenomicsNational Cancer Center‐Exploratory Oncology Research and Clinical Trial CenterTokyoJapan
| | - Kayoko Tao
- Department of Pediatric OncologyNational Cancer Center HospitalTokyoJapan
| | - Hourin Cho
- Department of Genetic Medicine and ServicesNational Cancer Center HospitalTokyoJapan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro‐OncologyNational Cancer Center HospitalTokyoJapan
| | - Ken Kato
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
- Department of Head and Neck, Esophageal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Yuichiro Ohe
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic OncologyNational Cancer Center HospitalTokyoJapan
| | | | - Chitose Ogawa
- Department of Pediatric OncologyNational Cancer Center HospitalTokyoJapan
| | - Kan Yonemori
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Noboru Yamamoto
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
36
|
Sinha S, Vegesna R, Mukherjee S, Kammula AV, Dhruba SR, Wu W, Kerr DL, Nair NU, Jones MG, Yosef N, Stroganov OV, Grishagin I, Aldape KD, Blakely CM, Jiang P, Thomas CJ, Benes CH, Bivona TG, Schäffer AA, Ruppin E. PERCEPTION predicts patient response and resistance to treatment using single-cell transcriptomics of their tumors. NATURE CANCER 2024; 5:938-952. [PMID: 38637658 DOI: 10.1038/s43018-024-00756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 03/08/2024] [Indexed: 04/20/2024]
Abstract
Tailoring optimal treatment for individual cancer patients remains a significant challenge. To address this issue, we developed PERCEPTION (PERsonalized Single-Cell Expression-Based Planning for Treatments In ONcology), a precision oncology computational pipeline. Our approach uses publicly available matched bulk and single-cell (sc) expression profiles from large-scale cell-line drug screens. These profiles help build treatment response models based on patients' sc-tumor transcriptomics. PERCEPTION demonstrates success in predicting responses to targeted therapies in cultured and patient-tumor-derived primary cells, as well as in two clinical trials for multiple myeloma and breast cancer. It also captures the resistance development in patients with lung cancer treated with tyrosine kinase inhibitors. PERCEPTION outperforms published state-of-the-art sc-based and bulk-based predictors in all clinical cohorts. PERCEPTION is accessible at https://github.com/ruppinlab/PERCEPTION . Our work, showcasing patient stratification using sc-expression profiles of their tumors, will encourage the adoption of sc-omics profiling in clinical settings, enhancing precision oncology tools based on sc-omics.
Collapse
Affiliation(s)
- Sanju Sinha
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA.
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA.
| | - Rahulsimham Vegesna
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Sumit Mukherjee
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Ashwin V Kammula
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
- University of Maryland, College Park, MD, USA
| | | | - Wei Wu
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - D Lucas Kerr
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nishanth Ulhas Nair
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Matthew G Jones
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
- Integrative Program in Quantitative Biology, University of California, San Francisco, San Francisco, CA, USA
- Whitehead Institute, Cambridge, MA, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
| | | | - Ivan Grishagin
- Rancho BioSciences, San Diego, CA, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Peng Jiang
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cyril H Benes
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub Investigator, San Francisco, CA, USA
| | | | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
37
|
Metzger P, Boerries M. [The collaborative project "Personalized medicine for oncology" (PM4Onco) as part of the Medical Informatics Initiative (MII)]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:668-675. [PMID: 38739266 PMCID: PMC11166753 DOI: 10.1007/s00103-024-03886-6] [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] [Accepted: 04/25/2024] [Indexed: 05/14/2024]
Abstract
The collaborative project Personalized Medicine for Oncology (PM4Onco) was launched in 2023 as part of the National Decade against Cancer (NKD) and is executed within the Medical Informatics Initiative (MII). Its aim is to establish a sustainable infrastructure for the integration and use of data from clinical and biomedical research and therefore combines the experience and preliminary work of all four consortia of the MII and the leading oncology centers in Germany. The data provided by PM4Onco will be prepared in a suitable form to support decision making in molecular tumor boards. This concept and infrastructure will be extended to 23 participating partner sites and thus improve access to targeted therapies based on clinical information and analysis of molecular genetic alterations in tumors at different stages of the disease. This will help to improve the treatment and prognosis of tumor diseases.Clinical cancer registries are involved in the project to improve data quality through standardized documentation routines. Clinical experts advise on the expansion of the core datasets for personalized medicine (PM). Information on quality of life and treatment outcomes reported by patients in questionnaires, which is rarely collected outside of clinical trials, will make a significant contribution. Patient representatives are involved from the onset to ensure that the important perspective of patients is taken into account in the decision-making process. PM4Onco thus creates an alliance between the MII, oncological centers of excellence, clinical cancer registries, young scientists, patients, and citizens to strengthen and advance PM in cancer therapy.
Collapse
Affiliation(s)
- Patrick Metzger
- Institut für Medizinische Bioinformatik und Systemmedizin (IBSM), Universitätsklinikum Freiburg, Medizinische Fakultät, Universität Freiburg, Breisacher Straße 153, 79110, Freiburg, Deutschland
| | - Melanie Boerries
- Institut für Medizinische Bioinformatik und Systemmedizin (IBSM), Universitätsklinikum Freiburg, Medizinische Fakultät, Universität Freiburg, Breisacher Straße 153, 79110, Freiburg, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort Freiburg, Kooperation zwischen DKFZ und Universitätsklinikum Freiburg, Universität Freiburg, Freiburg, Deutschland.
- Comprehensive Cancer Center Freiburg (CCCF), Universitätsklinikum Freiburg, Universität Freiburg, Freiburg, Deutschland.
| |
Collapse
|
38
|
Zhou Z, Lin T, Chen S, Zhang G, Xu Y, Zou H, Zhou A, Zhang Y, Weng S, Han X, Liu Z. Omics-based molecular classifications empowering in precision oncology. Cell Oncol (Dordr) 2024; 47:759-777. [PMID: 38294647 DOI: 10.1007/s13402-023-00912-8] [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] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine. CONCLUSION This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
39
|
Masucci M, Karlsson C, Blomqvist L, Ernberg I. Bridging the Divide: A Review on the Implementation of Personalized Cancer Medicine. J Pers Med 2024; 14:561. [PMID: 38929782 PMCID: PMC11204735 DOI: 10.3390/jpm14060561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
The shift towards personalized cancer medicine (PCM) represents a significant transformation in cancer care, emphasizing tailored treatments based on the genetic understanding of cancer at the cellular level. This review draws on recent literature to explore key factors influencing PCM implementation, highlighting the role of innovative leadership, interdisciplinary collaboration, and coordinated funding and regulatory strategies. Success in PCM relies on overcoming challenges such as integrating diverse medical disciplines, securing sustainable investment for shared infrastructures, and navigating complex regulatory landscapes. Effective leadership is crucial for fostering a culture of innovation and teamwork, essential for translating complex biological insights into personalized treatment strategies. The transition to PCM necessitates not only organizational adaptation but also the development of new professional roles and training programs, underscoring the need for a multidisciplinary approach and the importance of team science in overcoming the limitations of traditional medical paradigms. The conclusion underscores that PCM's success hinges on creating collaborative environments that support innovation, adaptability, and shared vision among all stakeholders involved in cancer care.
Collapse
Affiliation(s)
- Michele Masucci
- Department of Learning, Informatics, Management and Ethics (LIME), Karolinska Institutet, Tomtebodavägen 18B, 171 65 Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Claes Karlsson
- Department of Oncology-Pathology (Onc-Pat), Karolinska Institutet, Anna Steckséns gata 30A, D2:04, 171 65 Solna, Sweden;
| | - Lennart Blomqvist
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Anna Steckséns gata 53, 171 65 Solna, Sweden;
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
40
|
Moll M, Hecker J, Platig J, Zhang J, Ghosh AJ, Pratte KA, Wang RS, Hill D, Konigsberg IR, Chiles JW, Hersh CP, Castaldi PJ, Glass K, Dy JG, Sin DD, Tal-Singer R, Mouded M, Rennard SI, Anderson GP, Kinney GL, Bowler RP, Curtis JL, McDonald ML, Silverman EK, Hobbs BD, Cho MH. Polygenic and transcriptional risk scores identify chronic obstructive pulmonary disease subtypes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.20.24307621. [PMID: 38826461 PMCID: PMC11142287 DOI: 10.1101/2024.05.20.24307621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Rationale Genetic variants and gene expression predict risk of chronic obstructive pulmonary disease (COPD), but their effect on COPD heterogeneity is unclear. Objectives Define high-risk COPD subtypes using both genetics (polygenic risk score, PRS) and blood gene expression (transcriptional risk score, TRS) and assess differences in clinical and molecular characteristics. Methods We defined high-risk groups based on PRS and TRS quantiles by maximizing differences in protein biomarkers in a COPDGene training set and identified these groups in COPDGene and ECLIPSE test sets. We tested multivariable associations of subgroups with clinical outcomes and compared protein-protein interaction networks and drug repurposing analyses between high-risk groups. Measurements and Main Results We examined two high-risk omics-defined groups in non-overlapping test sets (n=1,133 NHW COPDGene, n=299 African American (AA) COPDGene, n=468 ECLIPSE). We defined "High activity" (low PRS/high TRS) and "severe risk" (high PRS/high TRS) subgroups. Participants in both subgroups had lower body-mass index (BMI), lower lung function, and alterations in metabolic, growth, and immune signaling processes compared to a low-risk (low PRS, low TRS) reference subgroup. "High activity" but not "severe risk" participants had greater prospective FEV 1 decline (COPDGene: -51 mL/year; ECLIPSE: - 40 mL/year) and their proteomic profiles were enriched in gene sets perturbed by treatment with 5-lipoxygenase inhibitors and angiotensin-converting enzyme (ACE) inhibitors. Conclusions Concomitant use of polygenic and transcriptional risk scores identified clinical and molecular heterogeneity amongst high-risk individuals. Proteomic and drug repurposing analysis identified subtype-specific enrichment for therapies and suggest prior drug repurposing failures may be explained by patient selection.
Collapse
|
41
|
Kim SY, Kim JH, Kim TY, Park SR, Yoon S, Lee S, Lee SH, Kim TM, Han SW, Kim HR, Yun H, Lee S, Kim J, Choi YL, Choi KS, Chae H, Ryu H, Lee GW, Zang DY, Ahn JB. Pragmatic nationwide master observational trial based on genomic alterations in advanced solid tumors: KOrean Precision Medicine Networking Group Study of MOlecular profiling guided therapy based on genomic alterations in advanced Solid tumors (KOSMOS)-II study protocol KCSG AL-22-09. BMC Cancer 2024; 24:574. [PMID: 38724991 PMCID: PMC11080169 DOI: 10.1186/s12885-024-12338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) has been introduced to many Korean institutions to support molecular diagnostics in cancer since 2017, when it became eligible for reimbursement by the National Health Insurance Service. However, the uptake of molecularly guided treatment (MGT) based on NGS results has been limited because of stringent regulations regarding prescriptions outside of approved indications, a lack of clinical trial opportunities, and limited access to molecular tumor boards (MTB) at most institutions. The KOSMOS-II study was designed to demonstrate the feasibility and effectiveness of MGT, informed by MTBs, using a nationwide precision medicine platform. METHODS The KOSMOS-II trial is a large-scale nationwide master observational study. It involves a framework for screening patients with metastatic solid tumors for actionable genetic alterations based on local NGS testing. It recommends MGT through a remote and centralized MTB meeting held biweekly. MGT can include one of the following options: Tier 1, the therapeutic use of investigational drugs targeting genetic alterations such as ALK, EGFR, ERBB2, BRAF, FH, ROS1, and RET, or those with high tumor mutational burden; Tier 2, comprising drugs with approved indications or those permitted for treatment outside of the indications approved by the Health Insurance Review and Assessment Service of Korea; Tier 3, involving clinical trials matching the genetic alterations recommended by the MTB. Given the anticipated proportion of patients receiving MGT in the range of 50% ± 3.25%, this study aims to enroll 1,000 patients. Patients must have progressed to one or more lines of therapy and undergone NGS before enrollment. DISCUSSION This pragmatic master protocol provides a mass-screening platform for rare genetic alterations and high-quality real-world data. Collateral clinical trials, translational studies, and clinico-genomic databases will contribute to generating evidence for drug repositioning and the development of new biomarkers. TRIAL REGISTRATION NCT05525858.
Collapse
Grants
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- Roche, Basel, Switzerland
- Lunit, Seoul, Republic of Korea
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sook Ryun Park
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Shinkyo Yoon
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Soohyeon Lee
- Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| | - Hongseok Yun
- Center for Genomic Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sejoon Lee
- Center for Precision Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jihun Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kui Son Choi
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, South Korea
| | - Heejung Chae
- Department of Internal Medicine, National Cancer Center, Goyang, South Korea
| | - Hyewon Ryu
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Gyeong-Won Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Dae Young Zang
- Department of Internal Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Joong Bae Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| |
Collapse
|
42
|
Lörsch AM, Jung J, Lange S, Pfarr N, Mogler C, Illert AL. [Personalized medicine in oncology]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:180-189. [PMID: 38568256 DOI: 10.1007/s00292-024-01315-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Due to the considerable technological progress in molecular and genetic diagnostics as well as increasing insights into the molecular pathogenesis of diseases, there has been a fundamental paradigm shift in the past two decades from a "one-size-fits-all approach" to personalized, molecularly informed treatment strategies. Personalized medicine or precision medicine focuses on the genetic, physiological, molecular, and biochemical differences between individuals and considers their effects on the development, prevention, and treatment of diseases. As a pioneer of personalized medicine, the field of oncology is particularly noteworthy, where personalized diagnostics and treatment have led to lasting change in the treatment of cancer patients in recent years. In this article, the significant change towards personalized treatment concepts, especially in the field of personalized oncology, will be discussed and examined in more detail.
Collapse
Affiliation(s)
- Alisa Martina Lörsch
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
| | - Johannes Jung
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
| | - Sebastian Lange
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - Nicole Pfarr
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Carolin Mogler
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Anna Lena Illert
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland.
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland.
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Klinik für Innere Medizin I, Abteilung für Hämatologie, Onkologie und Stammzelltransplantation, Universitätsklinikum Freiburg, Freiburg, Deutschland.
| |
Collapse
|
43
|
Walker M, Mayr EM, Koppermann ML, Terron A, Wagner Y, Kling C, Pfarr N. [Molecular pathological analysis through the ages]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:173-179. [PMID: 38619582 PMCID: PMC11045621 DOI: 10.1007/s00292-024-01326-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Molecular pathological examinations of tumor samples encompass a wide range of diagnostic analyses. Especially in recent years, numerous new biomarkers have come to the forefront-the analysis of which is crucial for therapy decisions. OBJECTIVES Within the field of molecular pathology, the demands of next generation sequencing (NGS)-based requirements have experienced massive growth in recent years. To meet this demand, methods are constantly being adapted and further developed. The following sections aim to illuminate how this trend arises and which analyses are gaining importance. METHODS The article provides an overview of the essential nucleic acid-based analysis techniques in the field of massive parallel sequencing. Terms such as DNA- and RNA-based techniques, as well as the associated analysis methods, are described, particularly with regard to their use in routine molecular pathological diagnostics. RESULTS The breadth of genomic sequencing has been steadily growing in recent years, particularly due to the increasing relevance of personalized medicine, along with the rising approvals of targeted therapeutics. This necessitates, among other things, the analysis of new biomarkers. The diagnostics as part of interdisciplinary molecular tumor boards (MTB) are now based on large gene panels (> 1 megabase). Furthermore, through the "Modellvorhaben Genomsequenzierung" § 64e, whole exome or whole genome sequencing has been made available for oncological patients. Given these developments, it is evident that future analyses will require the integration of additional omics fields, such as whole transcriptome analysis, epigenomics, and proteomics. CONCLUSION The challenges of personalized medicine along with the necessity of simultaneously assessing numerous new biomarkers require the implementation and execution of new techniques in molecular pathology whose complexity is steadily increasing.
Collapse
Affiliation(s)
- Maria Walker
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Eva-Maria Mayr
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Mai-Lan Koppermann
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Ana Terron
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Yoko Wagner
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Charlotte Kling
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Deutschland
| | - Nicole Pfarr
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland.
| |
Collapse
|
44
|
Gremke N, Rodepeter FR, Teply-Szymanski J, Griewing S, Boekhoff J, Stroh A, Tarawneh TS, Riera-Knorrenschild J, Balser C, Hattesohl A, Middeke M, Ross P, Litmeyer AS, Romey M, Stiewe T, Wündisch T, Neubauer A, Denkert C, Wagner U, Mack EKM. NGS-Guided Precision Oncology in Breast Cancer and Gynecological Tumors-A Retrospective Molecular Tumor Board Analysis. Cancers (Basel) 2024; 16:1561. [PMID: 38672643 PMCID: PMC11048446 DOI: 10.3390/cancers16081561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Precision oncology treatments are being applied more commonly in breast and gynecological oncology through the implementation of Molecular Tumor Boards (MTBs), but real-world clinical outcome data remain limited. Methods: A retrospective analysis was conducted in patients with breast cancer (BC) and gynecological malignancies referred to our center's MTB from 2018 to 2023. The analysis covered patient characteristics, next-generation sequencing (NGS) results, MTB recommendations, therapy received, and clinical outcomes. Results: Sixty-three patients (77.8%) had metastatic disease, and forty-four patients (54.3%) had previously undergone three or more lines of systemic treatment. Personalized treatment recommendations were provided to 50 patients (63.3%), while 29 (36.7%) had no actionable target. Ultimately, 23 patients (29.1%) underwent molecular-matched treatment (MMT). Commonly altered genes in patients with pan-gyn tumors (BC and gynecological malignancies) included TP53 (n = 42/81, 51.9%), PIK3CA (n = 18/81, 22.2%), BRCA1/2 (n = 10/81, 12.3%), and ARID1A (n = 9/81, 11.1%). Patients treated with MMT showed significantly prolonged progression-free survival (median PFS 5.5 vs. 3.5 months, p = 0.0014). Of all patients who underwent molecular profiling, 13.6% experienced a major clinical benefit (PFSr ≥ 1.3 and PR/SD ≥ 6 months) through precision oncology. Conclusions: NGS-guided precision oncology demonstrated improved clinical outcomes in a subgroup of patients with gynecological and breast cancers.
Collapse
Affiliation(s)
- Niklas Gremke
- Department of Gynecology, Gynecological Endocrinology and Oncology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (S.G.); (J.B.); (A.S.); (U.W.)
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany;
| | - Fiona R. Rodepeter
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Julia Teply-Szymanski
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Sebastian Griewing
- Department of Gynecology, Gynecological Endocrinology and Oncology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (S.G.); (J.B.); (A.S.); (U.W.)
| | - Jelena Boekhoff
- Department of Gynecology, Gynecological Endocrinology and Oncology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (S.G.); (J.B.); (A.S.); (U.W.)
| | - Alina Stroh
- Department of Gynecology, Gynecological Endocrinology and Oncology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (S.G.); (J.B.); (A.S.); (U.W.)
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany;
| | - Thomas S. Tarawneh
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (T.S.T.); (J.R.-K.); (P.R.); (A.N.); (E.K.M.M.)
| | - Jorge Riera-Knorrenschild
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (T.S.T.); (J.R.-K.); (P.R.); (A.N.); (E.K.M.M.)
| | - Christina Balser
- Practice for Internal Medicine, Hematology and Internal Oncology, 35043 Marburg, Germany;
| | - Akira Hattesohl
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Martin Middeke
- Comprehensive Cancer Center Marburg, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (M.M.); (T.W.)
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (T.S.T.); (J.R.-K.); (P.R.); (A.N.); (E.K.M.M.)
| | - Anne-Sophie Litmeyer
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Marcel Romey
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany;
| | - Thomas Wündisch
- Comprehensive Cancer Center Marburg, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (M.M.); (T.W.)
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (T.S.T.); (J.R.-K.); (P.R.); (A.N.); (E.K.M.M.)
| | - Carsten Denkert
- Institute of Pathology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (F.R.R.); (J.T.-S.); (A.H.); (A.-S.L.); (M.R.); (C.D.)
| | - Uwe Wagner
- Department of Gynecology, Gynecological Endocrinology and Oncology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (S.G.); (J.B.); (A.S.); (U.W.)
| | - Elisabeth K. M. Mack
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg Campus Marburg, Philipps-University, 35043 Marburg, Germany; (T.S.T.); (J.R.-K.); (P.R.); (A.N.); (E.K.M.M.)
| |
Collapse
|
45
|
Aydın E, Tokat ÜM, Özgü E, Adibi A, Tutar O, Kurzrock R, Demiray M. Navigating uncharted territory: a case report and literature review on the remarkable response to personalized crizotinib containing combinational therapy in a pazopanib refractory patient with novel alterations. Ther Adv Med Oncol 2024; 16:17588359241247023. [PMID: 38645422 PMCID: PMC11027594 DOI: 10.1177/17588359241247023] [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: 11/14/2023] [Accepted: 03/27/2024] [Indexed: 04/23/2024] Open
Abstract
This paper presents a patient with a novel Ig-like-III domain fibroblast growth factor receptor (FGFR2) alteration (W290_P307>C) along with CDKN2A/B alterations and a cadherin 1 (CDH1) alteration. Initial responsiveness to pazopanib monotherapy was encouraging, yet progression occurred after 7.5 months. Following progression, the molecular tumor board recommended a combination therapy approach comprising pazopanib, crizotinib, and palbociclib to target all of the changed pathways at the same time. Pazopanib was chosen to specifically target the FGFR2 alteration, while crizotinib was selected due to its potential synthetic lethality with the CDH1 alteration. In addition, the CDK4/6 inhibitor palbociclib was administered to address the CDKN2A/B alterations. The patient exhibited a remarkable and sustained response to this innovative combination. This case not only underscores the potential of tyrosine kinase inhibitors, exemplified by pazopanib, as a viable alternative for patients without access to pan-FGFR inhibitors, but it also emphasizes their efficacy beyond commonly detected point mutations and rearrangements. Notably, the outstanding response to combination therapy, including crizotinib, in a patient with a CDH1 alteration, further substantiates the preclinical evidence of synthetic lethality between crizotinib and CDH1 alterations. To our knowledge, this represents the first clinical evidence demonstrating the efficacy of crizotinib in a patient with a CDH1 alteration. Through careful dosage adjustments and consideration of individualized genomic information, this case exemplifies the power of personalized medicine in achieving favorable treatment outcomes.
Collapse
Affiliation(s)
- Esranur Aydın
- Demiray Precision Oncology Center, Medicana Health Group, Istanbul, Turkey
| | - Ünal Metin Tokat
- Demiray Precision Oncology Center, Medicana Health Group, Istanbul, Turkey
| | - Eylül Özgü
- Demiray Precision Oncology Center, Medicana Health Group, Istanbul, Turkey
| | - Ashkan Adibi
- Demiray Precision Oncology Center, Medicana Health Group, Istanbul, Turkey
- Division of Cancer Genetics, Department of Basic Oncology, Institute of Oncology, University of Istanbul, Istanbul, Turkey
| | - Onur Tutar
- Cerrahpaşa Faculty of Medicine, Department of Radiology, Istanbul University–Cerrahpasa, Istanbul, Turkey
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, 9200 W, Wisconsin Ave, Milwaukee, WI 53226, USA
| | - Mutlu Demiray
- Demiray Precision Oncology Center, Medicana Health Group, Küçükbakkalköy, Vedat Günyol Cd. No. 24, Ataşehir, Istanbul 34750, Turkey
| |
Collapse
|
46
|
Zhang L, Liu Q, Guo Y, Tian L, Chen K, Bai D, Yu H, Han X, Luo W, Feng T, Deng S, Xie G. DNA-based molecular classifiers for the profiling of gene expression signatures. J Nanobiotechnology 2024; 22:189. [PMID: 38632615 PMCID: PMC11025223 DOI: 10.1186/s12951-024-02445-0] [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: 09/28/2023] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
Although gene expression signatures offer tremendous potential in diseases diagnostic and prognostic, but massive gene expression signatures caused challenges for experimental detection and computational analysis in clinical setting. Here, we introduce a universal DNA-based molecular classifier for profiling gene expression signatures and generating immediate diagnostic outcomes. The molecular classifier begins with feature transformation, a modular and programmable strategy was used to capture relative relationships of low-concentration RNAs and convert them to general coding inputs. Then, competitive inhibition of the DNA catalytic reaction enables strict weight assignment for different inputs according to their importance, followed by summation, annihilation and reporting to accurately implement the mathematical model of the classifier. We validated the entire workflow by utilizing miRNA expression levels for the diagnosis of hepatocellular carcinoma (HCC) in clinical samples with an accuracy 85.7%. The results demonstrate the molecular classifier provides a universal solution to explore the correlation between gene expression patterns and disease diagnostics, monitoring, and prognosis, and supports personalized healthcare in primary care.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qian Liu
- Nuclear Medicine Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yongcan Guo
- Clinical Laboratory, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000, China
| | - Luyao Tian
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Kena Chen
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Bai
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hongyan Yu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaole Han
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wang Luo
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Tong Feng
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shixiong Deng
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Guoming Xie
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
47
|
Moussa P, Kurzrock R, Nishizaki D, Miyashita H, Lee S, Nikanjam M, Pabla S, Nesline MK, Ko H, Conroy JM, DePietro P, Sicklick JK, Kato S. Transcriptomic analysis of GITR and GITR ligand reveals cancer immune heterogeneity with implications for GITR targeting. Am J Cancer Res 2024; 14:1634-1648. [PMID: 38726288 PMCID: PMC11076267 DOI: 10.62347/eced5481] [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: 02/22/2024] [Accepted: 03/27/2024] [Indexed: 05/12/2024] Open
Abstract
Glucocorticoid-induced tumor necrosis factor related protein (GITR) is a transmembrane protein expressed mostly on CD25+CD4+ regulatory T-cells (Tregs) and upregulated on all T-cells upon activation. It is a T-cell co-stimulatory receptor and has demonstrated promising anti-tumor activity in pre-clinical studies. To date, however, the efficacy of GITR agonism has been discouraging in clinical trials. This study explores GITR and GITR ligand (GITR-L) ribonucleic acid (RNA) expression in solid tumors in an attempt to delineate causes for variable responses to GITR agonists. RNA expression levels of 514 patients with a variety of cancer types were normalized to internal housekeeping gene profiles and ranked as percentiles. 99/514 patients (19.3%) had high GITR expression (defined as ≥ 75th percentile). Breast and lung cancer had the highest proportion of patients with high GITR expression (39% and 35%, respectively). The expression of concomitant high GITR and low-moderate GITR-L expression (defined as <75th percentile) was present in 31% and 30% of patients with breast and lung cancer respectively. High GITR expression also showed a significant independent association with high RNA expression of other immune modulator proteins, namely, PD-L1 immunohistochemistry (IHC) ≥1 (odds ratio (OR) 2.15, P=0.008), CTLA4 (OR=2.17, P=0.05) and OX40 high RNA expression (OR=2.64, P=0.001). Overall, these results suggest that breast and lung cancer have a high proportion of patients with a GITR and GITR-L RNA expression profile that merits further investigation in GITR agonism studies. The association of high GITR expression with high CTLA4 and OX40 RNA expression, as well as positive PD-L1 IHC, provides a rationale for a combination approach targeting these specific immune modulator proteins in patients whose tumors show such co-expression.
Collapse
Affiliation(s)
- Peter Moussa
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology-Oncology, MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of WisconsinMilwaukee, WI, The United States
| | - Daisuke Nishizaki
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Hirotaka Miyashita
- Department of Medicine, Division of Hematology-Oncology, Dartmouth Cancer CenterLebanon, NH, The United States
| | - Suzanna Lee
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Mina Nikanjam
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | | | | | - Heidi Ko
- Labcorp OncologyDurham, NC, The United States
| | | | | | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, UC San DiegoSan Diego, CA, The United States
- Department of Pharmacology, UC San DiegoSan Diego, CA, The United States
- Structural and Functional Genomics Program, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Shumei Kato
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| |
Collapse
|
48
|
Danstrup CS, Andersen M, Lundbye-Christensen S, Sommer M, Lyhne NM. Survey of Danish Head and Neck Cancer Patients' Positions on Personalized Medicine, Gene Tests, and Personalized Follow-Up. J Pers Med 2024; 14:404. [PMID: 38673031 PMCID: PMC11051027 DOI: 10.3390/jpm14040404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
The field of personalized medicine (PM) has grown rapidly because of the "omics revolution", but PM may be difficult for patients to comprehend. This study sought to explore head and neck cancer (HNC) patients' positions and knowledge of PM, gene tests, and follow-up and to compare HNC patients' positions to a sample from a national Danish questionnaire. To do this, patients with prior HNC were invited to participate in a questionnaire. Initial interviews revealed a heterogenic understanding of PM between patients. A total of 226 patients were included in the survey and 177 patients with complete data were included for analysis. Most patients were more positive than negative towards gene tests and gene research (83% and 93%, respectively), but 72% had little or no knowledge of the subject. Almost all patients, 98%, were satisfied with their follow-up. Significantly more patients with HNC were positive towards gene research compared to a sample from a national Danish questionnaire (p < 0.001). Patients with HNC were positive towards gene tests and PM, but patients may not understand or comprehend the information given, and it is important to inform and educate patients and health professionals to establish common ground in PM.
Collapse
Affiliation(s)
- Christian Sander Danstrup
- Department of Otorhinolaryngology-Head and Neck Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Maria Andersen
- Department of Clinical Oncology, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Søren Lundbye-Christensen
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
- Research Data and Biostatistics, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Mia Sommer
- Department of Hematology, Aalborg University Hospital, 9000 Aalborg, Denmark
- School of Nursing, University College Northern Denmark, 9000 Aalborg, Denmark
| | - Nina Munk Lyhne
- Department of Otorhinolaryngology-Head and Neck Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
49
|
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
|
50
|
Hoo R, Chua KLM, Panda PK, Skanderup AJ, Tan DSW. Precision Endpoints for Contemporary Precision Oncology Trials. Cancer Discov 2024; 14:573-578. [PMID: 38571432 DOI: 10.1158/2159-8290.cd-24-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY Traditional endpoints such as progression-free survival and overall survival do not fully capture the pharmacologic and pharmacodynamic effects of a therapeutic intervention. Incorporating mechanism-driven biomarkers and validated surrogate proximal endpoints can provide orthogonal readouts of anti-tumor activity and delineate the relative contribution of treatment components on an individual level, highlighting the limitation of solely relying on aggregated readouts from clinical trials to facilitate go/no-go decisions for precision therapies.
Collapse
Affiliation(s)
- Regina Hoo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer and Therapeutics Research Laboratory, National Cancer Centre Singapore, Singapore
- Genome Institute of Singapore, Singapore
| | - Kevin L M Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Pankaj Kumar Panda
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | | | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer and Therapeutics Research Laboratory, National Cancer Centre Singapore, Singapore
- Genome Institute of Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| |
Collapse
|