1
|
García-Barberán V, Gómez Del Pulgar ME, Guamán HM, Benito-Martin A. The times they are AI-changing: AI-powered advances in the application of extracellular vesicles to liquid biopsy in breast cancer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:128-140. [PMID: 40206803 PMCID: PMC11977355 DOI: 10.20517/evcna.2024.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 01/03/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Artificial intelligence (AI) is revolutionizing scientific research by facilitating a paradigm shift in data analysis and discovery. This transformation is characterized by a fundamental change in scientific methods and concepts due to AI's ability to process vast datasets with unprecedented speed and accuracy. In breast cancer research, AI aids in early detection, prognosis, and personalized treatment strategies. Liquid biopsy, a noninvasive tool for detecting circulating tumor traits, could ideally benefit from AI's analytical capabilities, enhancing the detection of minimal residual disease and improving treatment monitoring. Extracellular vesicles (EVs), which are key elements in cell communication and cancer progression, could be analyzed with AI to identify disease-specific biomarkers. AI combined with EV analysis promises an enhancement in diagnosis precision, aiding in early detection and treatment monitoring. Studies show that AI can differentiate cancer types and predict drug efficacy, exemplifying its potential in personalized medicine. Overall, the integration of AI in biomedical research and clinical practice promises significant changes and advancements in diagnostics, personalized medicine-based approaches, and our understanding of complex diseases like cancer.
Collapse
Affiliation(s)
- Vanesa García-Barberán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - María Elena Gómez Del Pulgar
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Heidy M. Guamán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Alberto Benito-Martin
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio, Madrid 28691, Spain
| |
Collapse
|
2
|
Hoppe R, Winter S, Lo WY, Michailidou K, Bolla MK, Keeman R, Wang Q, Dennis J, Lush M, Kalari KR, Goetz MP, Wang L, Cairns J, Weinshilboum R, Shepherd L, Chen BE, Häberle L, Ruebner M, Beckmann MW, He W, Larson NL, Armasu SM, Schroth W, Chowbay B, Khor CC, Abubakar M, Antoniou AC, Brüning T, Castelao JE, Chang-Claude J, Nbcs Collaborators, Dörk T, Eccles DM, Figueroa JD, Gago-Dominguez M, García-Sáenz JA, Gündert M, Hack CC, Hamann U, Han S, Hooning MJ, Huebner H, Abctb Investigators, John EM, Ko YD, Kristensen VN, Linn S, Margolin S, Mavroudis D, Nevanlinna H, Neven P, Obi N, Park-Simon TW, Pylkäs K, Rashid MU, Romero A, Saloustros E, Sawyer EJ, Tapper WJ, Tomlinson I, Wendt C, Winqvist R, Dunning AM, Simard J, Hall P, Pharoah PDP, Schwab M, Couch FJ, Czene K, Fasching PA, Easton DF, Schmidt MK, Ingle JN, Brauch H. Lessons learned from a candidate gene study investigating aromatase inhibitor treatment outcome in breast cancer. NPJ Breast Cancer 2025; 11:18. [PMID: 39971965 PMCID: PMC11840073 DOI: 10.1038/s41523-025-00733-y] [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: 07/24/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
The role of germline genetics in adjuvant aromatase inhibitor (AI) treatment efficacy in ER-positive breast cancer is poorly understood. We employed a two-stage candidate gene approach to examine associations between survival endpoints and common germline variants in 753 endocrine resistance-related genes. For a discovery cohort, we screened the Breast Cancer Association Consortium database (n ≥ 90,000 cases) and retrieved 2789 AI-treated patients. Cox model-based analysis revealed 125 variants associated with overall, distant relapse-free, and relapse-free survival (p-value ≤ 1E-04). In validation analysis using five independent cohorts (n = 8857), none of the six selected candidates representing major linkage blocks at CELA2B/CASP9, NR1I2/GSK3B, LRP1B, and MIR143HG (CARMN) were validated. We discuss potential reasons for the failed validation and replication of published findings, including study/treatment heterogeneity and other limitations inherent to genomic treatment outcome studies. For the future, we envision prospective longitudinal studies with sufficiently long follow-up and endpoints that reflect the dynamic nature of endocrine resistance.
Collapse
Grants
- U19 GM061388 NIGMS NIH HHS
- 01KW0114 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- 110828 Deutsche Krebshilfe (German Cancer Aid)
- U10 CA077202 NCI NIH HHS
- 01KW9976/8 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- C1275/A15956 Cancer Research UK (CRUK)
- 2013PR044 Breast Cancer Campaign
- C12292/A11174 Cancer Research UK (CRUK)
- NMRC/CIRG/1423/2015 MOH | National Medical Research Council (NMRC)
- C5047/A15007 Cancer Research UK (CRUK)
- C1275/A11699 Cancer Research UK (CRUK)
- R01 CA192393 NCI NIH HHS
- R01 CA196648 NCI NIH HHS
- HEALTH-F2-2009-223175 EC | EC Seventh Framework Programm | FP7 Health (FP7-HEALTH - Specific Programme "Cooperation": Health)
- BMBF 01ZP0502 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- 2010PR62 Breast Cancer Campaign
- 01KW9977/0 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- R01 CA176785 NCI NIH HHS
- 01KH040 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- DFG Do761/15-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- C5047/A8384 Cancer Research UK (CRUK)
- PPRPGM-Nov20\100002 Cancer Research UK (CRUK)
- EXC 2180-390900677 Deutsche Forschungsgemeinschaft (German Research Foundation)
- 110826 Deutsche Krebshilfe (German Cancer Aid)
- CCS 015469 Canadian Cancer Society Research Institute (Société Canadienne du Cancer)
- 01KW9975/5 Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- 106332 Deutsche Krebshilfe (German Cancer Aid)
- C8197/A16565 Cancer Research UK (CRUK)
- C1287/A10710 Cancer Research UK (CRUK)
- C1287/A10118 Cancer Research UK (CRUK)
- R01 CA128978 NCI NIH HHS
- U19 CA148537 NCI NIH HHS
- C1275/A19187 Cancer Research UK (CRUK)
- R01 CA116167 NCI NIH HHS
- 70-2892-BR I Deutsche Krebshilfe (German Cancer Aid)
- P50 CA116201 NCI NIH HHS
- MOH-000377 MOH | National Medical Research Council (NMRC)
- C1287/A16563 Cancer Research UK (CRUK)
- C5047/A10692 Cancer Research UK (CRUK)
- U01 CA164920 NCI NIH HHS
- R35 CA253187 NCI NIH HHS
- 108419 Deutsche Krebshilfe (German Cancer Aid)
- U19 CA148112 NCI NIH HHS
- U19 CA148065 NCI NIH HHS
- SCHR 1323/2-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- 108253 Deutsche Krebshilfe (German Cancer Aid)
- C1275/C22524 Cancer Research UK (CRUK)
- C1281/A12014 Cancer Research UK (CRUK)
- MOH-000984 MOH | National Medical Research Council (NMRC)
- Robert Bosch Stiftung (Robert Bosch Foundation)
- Acción Estratégica de Salud del Instituto, de Salud Carlos III, (FIS PI12/02125/Cofinanciado), Acción Estratégica de Salud del Instituto, de Salud Carlos III, (FEDER PI17/00918/Cofinanciado, FEDER), Acción Estratégica de Salud del Instituto, de Salud Carlos III, (FIS Intrasalud PI13/01136), Programa Grupos Emergentes, Cancer, Genetics Unit, Instituto de Investigacion, Biomedica Galicia Sur. Xerencia de, Xestion Integrada de Vigo-SERGAS,, Instituto de Salud Carlos III, (10CSA012E), Consellería de Industria Programa, Sectorial de Investigación Aplicada,, PEME I + D e I + D Suma del Plan, Gallego de Investigación, Desarrollo e, Innovación Tecnológica de la Consellería, de Industria de la Xunta de Galicia, (EC11-192)
- Dutch Cancer Society (DDHK 2004-3124) Dutch Cancer Society (DDHK 2009-4318)
- NCI/NIH, (U01CA164920)
- Finnish Cancer Foundation, the Academy of Finland, (250083), (122715), (251314)
- Genome Canada and the Canadian, Institutes of Health Research, (GPH-129344) Genome Québec, (PSRSIIRI-701)
- NIH, (R35CA253187), (R01CA192393), (R01CA116167), (R01CA176785) NIH Specialized Program of Research, Excellence (SPORE), (P50CA116201)
- University Hospital Erlangen, (UISGE-005/2018)
- Dutch Cancer Society, (NKI 2007-3839) Dutch Cancer Society, (NKI 2009 4363)
- NIH, (U19 GM61388) NIH, (P50CA116201) NIH, (U10CA77202) NIH, (R01CA196648)
Collapse
Affiliation(s)
- Reiner Hoppe
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.
- University of Tübingen, Tübingen, Germany.
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Wing-Yee Lo
- Department of Clinical Pathology, Centre for Cancer Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Manjeet K Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Renske Keeman
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Michael Lush
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Junmei Cairns
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Lois Shepherd
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Bingshu E Chen
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Wei He
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nicole L Larson
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Sebastian M Armasu
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Werner Schroth
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Balram Chowbay
- Laboratory of Clinical Pharmacology, Division of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Centre for Clinician Scientist Development, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology & Research (A*STAR), Singapore, Singapore
| | - Chiea Chuen Khor
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum, Bochum, Germany
| | - Jose E Castelao
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS) Foundation, Complejo Hospitalario Universitario de Santiago, SERGAS, Vigo, Spain
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nbcs Collaborators
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Research, Vestre Viken Hospital, Drammen, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital - Radiumhospitalet, Oslo, Norway
- Department of Oncology, Division of Surgery and Cancer and Transplantation Medicine, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Oslo Breast Cancer Research Consortium, Oslo University Hospital, Oslo, Norway
- Department of Community Medicine, The Arctic University of Norway, Tromsø, Norway
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Diana M Eccles
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Manuela Gago-Dominguez
- Cancer Genetics and Epidemiology Group, Genomic Medicine Group, Fundación Pública Galega de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Complejo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain
| | - José A García-Sáenz
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Melanie Gündert
- Molecular Epidemiology Group, C080, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Biology of Breast Cancer, University Womens Clinic Heidelberg, University of Heidelberg, Heidelberg, Germany
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Carolin C Hack
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sileny Han
- Leuven Multidisciplinary Breast Center, Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Abctb Investigators
- Australian Breast Cancer Tissue Bank, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Esther M John
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter GmbH Bonn, Johanniter Krankenhaus, Bonn, Germany
| | - Vessela N Kristensen
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sabine Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sara Margolin
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion, Greece
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Patrick Neven
- Leuven Multidisciplinary Breast Center, Department of Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Nadia Obi
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Occupational and Maritime Medicine (ZfAM), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, Medical Research Centre, University of Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre Oulu, Oulu, Finland
| | - Muhammad U Rashid
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Basic Sciences, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH & RC), Lahore, Pakistan
| | - Atocha Romero
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Elinor J Sawyer
- School of Cancer & Pharmaceutical Sciences, Comprehensive Cancer Centre, Guy's Campus, King's College London, London, UK
| | | | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, UK
| | - Camilla Wendt
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit, Biocenter Oulu, Medical Research Centre, University of Oulu, Oulu, Finland
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Québec City, Québec, Canada
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Paul D P Pharoah
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Zhang Y, Yuan X. Minimal residue disease detection in early-stage breast cancer: a review. Mol Biol Rep 2025; 52:106. [PMID: 39777588 DOI: 10.1007/s11033-024-10198-0] [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: 11/20/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Over the past five years, circulating tumor DNA (ctDNA) testing has emerged as a game-changer in cancer research, serving as a less invasive and highly sensitive method to monitor tumor dynamics. CtDNA testing has a wide range of potential applications in breast cancer (BC) management, including diagnosis, monitoring treatment responses, identifying resistance mutations, predicting prognosis, and detecting future relapses. In this review, we focus on the prognostic and predictive value of ctDNA testing for BC in both neoadjuvant and adjuvant settings. We also examine the rationale behind mainstream minimal residue disease (MRD) tracking methods and highlight key considerations for successful MRD testing. Clinical evidence has shown that ctDNA-based MRD testing can accurately detect molecular relapse 8-12 months before clinical relapse in early-stage BC. Compared to advanced-stage BC, detecting ctDNA in early-stage BC is more challenging and requires ultra-sensitive testing methods due to the low levels of ctDNA released into the bloodstream, particularly in post-surgical settings, after effective neoadjuvant chemotherapy, and in late adjuvant settings that require longer follow-up. Therefore, future efforts are needed to generate additional clinical evidence in these settings to support the clinical utility and widespread adoption of ctDNA-based MRD testing.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Art and Science, Northeast Agricultural University, Changjiang Road No. 600, Harbin, 150030, China
- Shuwen Biotech Co., Ltd., Moganshan National High tech Zone, Building 3, No. 333, Changhong Middle Street, Deqing, China
| | - Xiaoying Yuan
- Shuwen Biotech Co., Ltd., Moganshan National High tech Zone, Building 3, No. 333, Changhong Middle Street, Deqing, China.
| |
Collapse
|
4
|
Cortes-Mejia NA, Lillemoe HA, Cata JP. Return to Intended Oncological Therapy: State of the Art and Perspectives. Curr Oncol Rep 2024; 26:1420-1430. [PMID: 39320576 DOI: 10.1007/s11912-024-01594-7] [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] [Accepted: 08/06/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE OF THE REVIEW Despite advances in surgical procedures, cancer recurrence still affects a substantial proportion of patients for whom surgery is considered a curative therapy. This review aims to provide a comprehensive overview of RIOT, addressing its definition, influencing factors, and clinical implications. FINDINGS RIOT can be defined as a continuous variable as the time from surgery to initiation of adjuvant therapies or categorically as whether patients can successfully receive adjuvant therapies or not. Factors influencing RIOT are age, sex, socioeconomic status, access to healthcare, physical performance and comorbidities, and quality of anesthesia and surgical care. Adjuvant therapies such as chemotherapy, radiotherapy, and immunotherapy are often administered to reduce the risk of recurrence after surgery and improve survival. Return to intended oncologic therapy (RIOT) has emerged as a promising outcome metric reflecting patients' functional recovery after surgery and their ability to receive adjuvant therapies.
Collapse
Affiliation(s)
- Nicolas A Cortes-Mejia
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Heather A Lillemoe
- Department of Breast Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA.
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.
- Department of Pain Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Chen JH, Addanki S, Roy D, Bassett R, Kalashnikova E, Spickard E, Kuerer HM, Meas S, Sarli VN, Korkut A, White JB, Rauch GM, Tripathy D, Arun BK, Barcenas CH, Yam C, Sethi H, Rodriguez AA, Liu MC, Moulder SL, Lucci A. Monitoring response to neoadjuvant chemotherapy in triple negative breast cancer using circulating tumor DNA. BMC Cancer 2024; 24:1016. [PMID: 39148033 PMCID: PMC11328413 DOI: 10.1186/s12885-024-12689-6] [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] [Received: 03/15/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an aggressive subtype with poor prognosis. We aimed to determine whether circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) could predict response and long-term outcomes to neoadjuvant chemotherapy (NAC). METHODS Patients with TNBC were enrolled between 2017-2021 at The University of Texas MD Anderson Cancer Center (Houston, TX). Serial plasma samples were collected at four timepoints: pre-NAC (baseline), 12-weeks after NAC (mid-NAC), after NAC/prior to surgery (post-NAC), and one-year after surgery. ctDNA was quantified using a tumor-informed ctDNA assay (SignateraTM, Natera, Inc.) and CTC enumeration using CellSearch. Wilcoxon and Fisher's exact tests were used for comparisons between groups and Kaplan-Meier analysis used for survival outcomes. RESULTS In total, 37 patients were enrolled. The mean age was 50 and majority of patients had invasive ductal carcinoma (34, 91.9%) with clinical T2, (25, 67.6%) node-negative disease (21, 56.8%). Baseline ctDNA was detected in 90% (27/30) of patients, of whom 70.4% (19/27) achieved ctDNA clearance by mid-NAC. ctDNA clearance at mid-NAC was significantly associated with pathologic complete response (p = 0.02), whereas CTC clearance was not (p = 0.52). There were no differences in overall survival (OS) and recurrence-free survival (RFS) with positive baseline ctDNA and CTC. However, positive ctDNA at mid-NAC was significantly associated with worse OS and RFS (p = 0.0002 and p = 0.0034, respectively). CONCLUSIONS Early clearance of ctDNA served as a predictive and prognostic marker in TNBC. Personalized ctDNA monitoring during NAC may help predict response and guide treatment.
Collapse
Affiliation(s)
- Jennifer H Chen
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Sridevi Addanki
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Dhruvajyoti Roy
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Roland Bassett
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | - Henry M Kuerer
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Salyna Meas
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Vanessa N Sarli
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jason B White
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Gaiane M Rauch
- Abdominal Imaging Department, MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Banu K Arun
- Breast Medical Oncology and Clinical Cancer Genetics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Carlos H Barcenas
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Clinton Yam
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Stacy L Moulder
- Medical Oncology, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US.
| |
Collapse
|
6
|
Elliott MJ, Shen S, Lam DL, Brown T, Lawson MB, Iyengar NM, Cescon DW. Enhancing Early-Stage Breast Cancer Survivorship: Evidence-Based Strategies, Surveillance Testing, and Imaging Guidelines. Am Soc Clin Oncol Educ Book 2024; 44:e432564. [PMID: 38815189 DOI: 10.1200/edbk_432564] [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: 06/01/2024]
Abstract
Addressing the challenges of survivorship necessitates a comprehensive, patient-centered approach, focusing on mitigating risk through lifestyle modification, identifying distant recurrence, and optimization of breast imaging. This article will discuss the current and emerging clinical strategies for the survivorship period, advocating a multidisciplinary and comprehensive approach. In this manner, early-stage breast cancer survivors are empowered to navigate their journey with enhanced knowledge, facilitating a transition to life beyond cancer.
Collapse
Affiliation(s)
- Mitchell J Elliott
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sherry Shen
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Diana L Lam
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | - Thelma Brown
- University of Alabama at Birmingham, Birmingham, AL
| | - Marissa B Lawson
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | | | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
7
|
Karihtala P, Leivonen SK, Puistola U, Urpilainen E, Jääskeläinen A, Leppä S, Jukkola A. Serum protein profiling reveals an inflammation signature as a predictor of early breast cancer survival. Breast Cancer Res 2024; 26:61. [PMID: 38594742 PMCID: PMC11005292 DOI: 10.1186/s13058-024-01812-x] [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: 10/21/2023] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Breast cancers exhibit considerable heterogeneity in their biology, immunology, and prognosis. Currently, no validated, serum protein-based tools are available to evaluate the prognosis of patients with early breast cancer. METHODS The study population consisted of 521 early-stage breast cancer patients with a median follow-up of 8.9 years. Additionally, 61 patients with breast fibroadenoma or atypical ductal hyperplasia were included as controls. We used a proximity extension assay to measure the preoperative serum levels of 92 proteins associated with inflammatory and immune response processes. The invasive cancers were randomly split into discovery (n = 413) and validation (n = 108) cohorts for the statistical analyses. RESULTS Using LASSO regression, we identified a nine-protein signature (CCL8, CCL23, CCL28, CSCL10, S100A12, IL10, IL10RB, STAMPB2, and TNFβ) that predicted various survival endpoints more accurately than traditional prognostic factors. In the time-dependent analyses, the prognostic power of the model remained rather stable over time. We also developed and validated a 17-protein model with the potential to differentiate benign breast lesions from malignant lesions (Wilcoxon p < 2.2*10- 16; AUC 0.94). CONCLUSIONS Inflammation and immunity-related serum proteins have the potential to rise above the classical prognostic factors of early-stage breast cancer. They may also help to distinguish benign from malignant breast lesions.
Collapse
Affiliation(s)
- Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, P.O. Box 180, Helsinki, FI-00029, Finland.
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Suvi-Katri Leivonen
- Applied Tumor Genomics, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynecology, Medical Research Center, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
| | - Elina Urpilainen
- Department of Obstetrics and Gynecology, Medical Research Center, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, Oulu, Finland
| | - Anniina Jääskeläinen
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, P.O. Box 180, Helsinki, FI-00029, Finland
- Applied Tumor Genomics, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Arja Jukkola
- Department of Oncology, Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University, Tampere, Finland
| |
Collapse
|
8
|
Chakraborty A, Mitra A, Sahu S, Tawate M, Lad S, Kamaldeep, Rakshit S, Upadhye Bannore T, Gaikwad S, Dhotre G, Ray MK, Damle A, Basu S, Banerjee S. Intricacies in the Preparation of Patient Doses of [ 177Lu]Lu-Rituximab and [ 177Lu]Lu-Trastuzumab Using Low Specific Activity [ 177Lu]LuCl 3: Methodological Aspects. Mol Imaging Biol 2024; 26:61-80. [PMID: 37673943 DOI: 10.1007/s11307-023-01846-1] [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: 12/28/2022] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023]
Abstract
The development of humanized monoclonal antibodies (MAbs) with Lutetium-177 ([177Lu]Lu3+) has brought a paradigm shift in the arena of targeted therapy of various cancers. [177Lu]Lu-DOTA-Rituximab and [177Lu]Lu-DOTA-Trastuzumab have gained prominence due to their improved therapeutic efficacy in the treatment of lymphoma and breast cancer. The clinical dose formulation of these radiolabeled MAbs, using low specific activity [177Lu]LuCl3, requires extensive optimization of the radiolabeling protocol. The present study merits the development of a single protocol which has been optimized for conjugation of Rituximab and Trastuzumab with p-NCS-benzyl-DOTA and further radiolabeling these immunoconjugates (ICs) with low specific activity [177Lu]LuCl3. Herein, we report a consistent and reproducible protocol for clinical dose formulations of [177Lu]Lu-DOTA-Rituximab and [177Lu]Lu-DOTA-Trastuzumab (~9.25 GBq each, equivalent to ~2 patient doses) with radiochemical yield (RCY) between 84 and 86% and radiochemical purities (RCP) >99%. The in vitro stabilities of both these radioimmunoconjugates (RICs) were retained up to 120 h post-radiolabeling, upon storage with L-ascorbic acid as stabilizer (concentration: ~ 220-240 μg/37MBq) at -20 °C. The ready-to-use formulation of clinical doses[177Lu]Lu-DOTA-Rituximab and [177Lu]Lu-DOTA-Trastuzumab has been successfully achieved by employing a single optimized protocol. While [177Lu]Lu-DOTA-Rituximab has exhibited a high degree of localization in retroperitoneal nodal mass of refractory lymphoma patient, high uptake of [177Lu]Lu-DOTA-Trastuzumab has been observed in metastatic breast carcinoma patient with multiple skeletal metastases.
Collapse
Affiliation(s)
- Avik Chakraborty
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Arpit Mitra
- Radiopharmaceuticals Laboratory, Board of Radiation and Isotope Technology, Vashi, Navi Mumbai, India
| | - Sudeep Sahu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
| | - Megha Tawate
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Sangita Lad
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
| | - Kamaldeep
- Homi Bhabha National Institute, Mumbai, India
- Health Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
| | | | - Sujay Gaikwad
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
| | - Geetanjali Dhotre
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Mukti Kanta Ray
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Archana Damle
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Sharmila Banerjee
- Homi Bhabha National Institute, Mumbai, India.
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410 210, India.
| |
Collapse
|
9
|
Medford AJ, Moy B, Spring LM, Hurvitz SA, Turner NC, Bardia A. Molecular Residual Disease in Breast Cancer: Detection and Therapeutic Interception. Clin Cancer Res 2023; 29:4540-4548. [PMID: 37477704 DOI: 10.1158/1078-0432.ccr-23-0757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
Breast cancer remains a leading cause of cancer-related death in women despite screening and therapeutic advances. Early detection allows for resection of local disease; however, patients can develop metastatic recurrences years after curative treatment. There is no reliable blood-based monitoring after curative therapy, and radiographic evaluation for metastatic disease is performed only in response to symptoms. Advances in circulating tumor DNA (ctDNA) assays have allowed for a potential option for blood-based monitoring. The detection of ctDNA in the absence of overt metastasis or recurrent disease indicates molecular evidence of cancer, defined as molecular residual disease (MRD). Multiple studies have shown that MRD detection is strongly associated with disease recurrence, with a lead time prior to clinical evidence of recurrence of many months. Importantly, it is still unclear whether treatment changes in response to ctDNA detection will improve outcomes. There are currently ongoing trials evaluating the efficacy of therapy escalation in the setting of MRD, and these studies are being conducted in all major breast cancer subtypes. Additional therapies under study include CDK4/6 inhibitors, PARP inhibitors, HER2-targeted therapies, and immunotherapy. This review will summarize the underlying scientific principles of various MRD assays, their known prognostic roles in early breast cancer, and the ongoing clinical trials assessing the efficacy of therapy escalation in the setting of MRD.
Collapse
Affiliation(s)
- Arielle J Medford
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Laura M Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Sara A Hurvitz
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Nicholas C Turner
- The Royal Marsden NHS Foundation Trust, Breast Cancer Now Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Abstract
The pattern of delayed recurrence in a subset of breast cancer patients has long been explained by a model that incorporates a variable period of cellular or tumor mass dormancy prior to disease relapse. In this review, we critically evaluate existing data to develop a framework for inferring the existence of dormancy in clinical contexts of breast cancer. We integrate these clinical data with rapidly evolving mechanistic insights into breast cancer dormancy derived from a broad array of genetically engineered mouse models as well as experimental models of metastasis. Finally, we propose actionable interventions and discuss ongoing clinical trials that translate the wealth of knowledge gained in the laboratory to the long-term clinical management of patients at a high risk of developing recurrence.
Collapse
Affiliation(s)
- Erica Dalla
- Division of Hematology and Oncology, Department of Medicine and Department of Otolaryngology, Department of Oncological Sciences, Black Family Stem Cell Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Amulya Sreekumar
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Julio A Aguirre-Ghiso
- Department of Cell Biology, Department of Oncology, Cancer Dormancy and Tumor Microenvironment Institute, Montefiore Einstein Cancer Center, Gruss Lipper Biophotonics Center, Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Lewis A Chodosh
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Medicine, Abramson Cancer Center, and 2-PREVENT Translational Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
11
|
Sposito M, Belluomini L, Pontolillo L, Tregnago D, Trestini I, Insolda J, Avancini A, Milella M, Bria E, Carbognin L, Pilotto S. Adjuvant Targeted Therapy in Solid Cancers: Pioneers and New Glories. J Pers Med 2023; 13:1427. [PMID: 37888038 PMCID: PMC10608226 DOI: 10.3390/jpm13101427] [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: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Targeted therapy (TT) has revolutionized cancer treatment, successfully applied in various settings. Adjuvant TT in resected early-stage gastrointestinal stromal tumors (GIST), melanoma, non-small cell lung cancer (NSCLC), and breast cancer has led to practice-changing achievements. In particular, standard treatments include BRAF inhibitors for melanoma, osimertinib for NSCLC, hormone therapy or HER2 TT for breast cancer, and imatinib for GIST. Despite the undeniable benefit derived from adjuvant TT, the optimal duration of TT and the appropriate managing of the relapse remain open questions. Furthermore, neoadjuvant TT is emerging as valuable, particularly in breast cancer, and ongoing studies evaluate TT in the perioperative setting for early-stage NSCLC. In this review, we aim to collect and describe the large amount of data available in the literature about adjuvant TT across different histologies, focusing on epidemiology, major advances, and future directions.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Letizia Pontolillo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (L.P.); (E.B.)
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Daniela Tregnago
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Ilaria Trestini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Jessica Insolda
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Alice Avancini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Michele Milella
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (L.P.); (E.B.)
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Luisa Carbognin
- Gynecology Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Roma, Italy;
| | - Sara Pilotto
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| |
Collapse
|
12
|
Abstract
The steady, incremental improvements in outcomes for both early-stage and advanced breast cancer patients are, in large part, attributable to the success of novel systemic therapies. In this review, we discuss key conceptual paradigms that have underpinned this success including (1) targeting the driver: the identification and targeting of major oncoproteins in breast cancers; (2) targeting the lineage pathway: inhibition of those pathways that drive normal mammary epithelial cell proliferation that retain importance in cancer; (3) targeting precisely: the application of molecular classifiers to refine therapy selection for specific cancers, and of antibody-drug conjugates to pinpoint tumor and tumor promoting cells for eradication; and (4) exploiting synthetic lethality: leveraging unique vulnerabilities that cancer-specific molecular alterations induce. We describe promising examples of novel therapies that have been discovered within each of these paradigms and suggest how future drug development efforts might benefit from the continued application of these principles.
Collapse
Affiliation(s)
- Shom Goel
- Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3010, Australia
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Weill Cornell Medicine, New York, New York 10021, USA
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| |
Collapse
|
13
|
Kim MS, Wu HG, Sung MW, Kwon TK. Long-term impact of smoking cessation on new glottic cancer events in patients with early glottic cancer. ACTA OTORHINOLARYNGOLOGICA ITALICA : ORGANO UFFICIALE DELLA SOCIETA ITALIANA DI OTORINOLARINGOLOGIA E CHIRURGIA CERVICO-FACCIALE 2022; 42:525-530. [PMID: 36654518 PMCID: PMC9853108 DOI: 10.14639/0392-100x-n1917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 06/13/2022] [Indexed: 01/18/2023]
Abstract
Objective Patients with early glottic cancer sometimes exhibit new glottic cancer events after 5 years. This study aimed to analyse the patterns and risk factors of new glottic cancer events in patients with early glottic cancer 5 years after initial treatment. Methods In total, 209 patients were included in this study. Age, sex, T stage, anterior commissure involvement, smoking pattern and treatment modality were retrospectively analysed. Results The median follow-up was 91 (range, 60-266) months. The median time for the occurrence of new glottic cancer events was 97 (range, 61-199) months. New glottic cancer events occurred 5 years after initial treatment in 16 (7.6%) patients, among whom 12 (75.0%) had new glottic cancer event lesions overlapping with initial lesions. Smoking cessation after treatment was significantly correlated with fewer new glottic cancer events after 5 years. Conclusions New glottic cancer events occurring 5 years after initial treatment in patients with early glottic cancer are not negligible. In particular, if smoking is continued after treatment, these patients can experience new glottic cancer events even after 5 years.
Collapse
Affiliation(s)
- Min-Su Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Republic of Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myung-Whun Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tack-Kyun Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea,Correspondence Tack-Kyun Kwon Department of Otorhinolaryngology-Head-and-Neck Surgery, Seoul National University College of Medicine, Boramae Medical Center, Room No 11221, 11th floor, Hangbok Building, Boramae Medical Center 20, Boramae-ro 5-gil, Dongjak-gu, Seoul 07061, Republic of Korea E-mail:
| |
Collapse
|
14
|
Lipsyc-Sharf M, de Bruin EC, Santos K, McEwen R, Stetson D, Patel A, Kirkner GJ, Hughes ME, Tolaney SM, Partridge AH, Krop IE, Knape C, Feger U, Marsico G, Howarth K, Winer EP, Lin NU, Parsons HA. Circulating Tumor DNA and Late Recurrence in High-Risk Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer. J Clin Oncol 2022; 40:2408-2419. [PMID: 35658506 PMCID: PMC9467679 DOI: 10.1200/jco.22.00908] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To examine the prevalence and dynamics of circulating tumor DNA (ctDNA) and its association with metastatic recurrence in patients with high-risk early-stage hormone receptor-positive breast cancer (HR+ BC) more than 5 years from diagnosis. METHODS We enrolled 103 patients with high-risk stage II-III HR+ BC diagnosed more than 5 years prior without clinical evidence of recurrence. We performed whole-exome sequencing (WES) on primary tumor tissue to identify somatic mutations tracked via a personalized, tumor-informed ctDNA test to detect minimal residual disease (MRD). We collected plasma at the time of consent and at routine visits every 6-12 months. Patients were followed for clinical recurrence. RESULTS In total, 85 of 103 patients had sufficient tumor tissue; of them, 83 of 85 (97.6%) patients had successful whole-exome sequencing. Personalized ctDNA assays were designed targeting a median of 36 variants to test 219 plasma samples. The median time from diagnosis to first sample was 8.4 years. The median follow-up was 10.4 years from diagnosis and 2.0 years from first sample. The median number of plasma samples per patient was two. Eight patients (10%) had positive MRD testing at any time point. Six patients (7.2%) developed distant metastatic recurrence, all of whom were MRD-positive before overt clinical recurrence, with median ctDNA lead time of 12.4 months. MRD was not identified in one patient (1.2%) with local recurrence. Two of eight MRD-positive patients had not had clinical recurrence at last follow-up. CONCLUSION In this prospective study, in patients with high-risk HR+ BC in the late adjuvant setting, ctDNA was identified a median of 1 year before all cases of distant metastasis. Future studies will determine if ctDNA-guided intervention in patients with HR+ BC can alter clinical outcomes.
Collapse
Affiliation(s)
- Marla Lipsyc-Sharf
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | | | | | | | | | - Ashka Patel
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Ann H Partridge
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Ian E Krop
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA.,Present affiliation: Yale University, New Haven, CT
| | | | - Ute Feger
- Inivata Inc, Research Triangle Park, NC
| | | | | | - Eric P Winer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA.,Present affiliation: Yale University, New Haven, CT
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Heather A Parsons
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Cescon DW, Kalinsky K, DeMichele AM. Can a Late Interception by Circulating Tumor DNA Deliver a Win in Estrogen Receptor-Positive Early Breast Cancer? J Clin Oncol 2022; 40:2395-2397. [PMID: 35658519 DOI: 10.1200/jco.22.01026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- David W Cescon
- Department of Medical Oncology, Princess Margaret Cancer Center, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA
| | | |
Collapse
|
16
|
Thomas A, Parsons HA, Smith KL. Late Recurrence Following Early Breast Cancer. J Clin Oncol 2022; 40:1400-1406. [PMID: 35239445 PMCID: PMC9061154 DOI: 10.1200/jco.22.00167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in the Journal of Clinical Oncology, to patients seen in their own clinical practice.
Collapse
Affiliation(s)
- Alexandra Thomas
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | | | - Karen Lisa Smith
- Women's Malignancies Disease Group, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|