1
|
Dang M, Schritz A, Goncharenko N, Berchem G. Impact of molecular diagnostics and targeted cancer therapy on patient outcomes (MODIFY): a retrospective study of the implementation of precision oncology. Mol Oncol 2024. [PMID: 39661533 DOI: 10.1002/1878-0261.13785] [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: 07/10/2024] [Revised: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
High-throughput genomic analyses are being implemented in clinical practice. MODIFY is a retrospective study of the first introduction of genomic profiling and molecular tumor boards in the country of Luxembourg. The primary objective was to assess whether patients derived a clinical benefit by measuring the percentage of patients who presented a progression-free survival (PFS) on matched therapy (PFS2) 1.3-fold longer than PFS on previous therapy (PFS1). A total of 94 patients were included. In total, 45 patients (53.57% of patients with successful next-generation sequencing [NGS] analysis) were found to have an actionable mutation. Of these, 11 patients received the treatment recommended by the molecular tumor board, another 12 received best-supportive care, and 20 were treated with conventional therapy. PFS2 and PFS1 data were available for eight patients. The PFS2/PFS1 ratio was ≥ -1.3 in 62.5% (n = 5/8; CI [30.38, 86.51]) of patients; three patients showed a partial response, and median overall survival (OS) was 7.3 months. Although the examined population was small, this study further supports evidence indicating that patients with advanced cancer benefit from molecular profiling and targeted therapy.
Collapse
Affiliation(s)
- Michaël Dang
- Department of Oncology, Centre Hospitalier de Luxembourg (CHL), Luxembourg
- University of Luxembourg (Uni.lu), Luxembourg
| | - Anna Schritz
- Luxembourg Institute of Health (LIH), Luxembourg
| | | | - Guy Berchem
- Department of Oncology, Centre Hospitalier de Luxembourg (CHL), Luxembourg
- University of Luxembourg (Uni.lu), Luxembourg
- Institut National du Cancer (INC), Luxembourg, Luxembourg
| |
Collapse
|
2
|
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
|
3
|
van Berge Henegouwen JM, Zeverijn LJ, Geurts BS, Hoes LR, van der Wijngaart H, van der Noort V, Huitema ADR, de Vos FYF, Grünberg K, Bloemendal HJ, Verheul HMW, Voest EE, Gelderblom H. Maximizing Treatment Opportunities: Assessing Protocol Waivers' Impact on Safety and Outcome in the Drug Rediscovery Protocol. Clin Cancer Res 2024; 30:3937-3943. [PMID: 38926908 DOI: 10.1158/1078-0432.ccr-23-3917] [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: 12/14/2023] [Revised: 02/12/2024] [Accepted: 03/29/2024] [Indexed: 06/28/2024]
Abstract
PURPOSE Although eligibility criteria are essential in trial design, overly restrictive criteria contribute to low accrual and limited generalizability. To enhance trial inclusivity, there has been growing interest in broadening eligibility criteria, especially for patients with advanced or treatment-refractory disease. Yet, the impact on patient safety remains uncertain. In the Drug Rediscovery Protocol (DRUP), protocol exceptions are frequently requested and occasionally granted. Here we describe the impact of these waivers on treatment safety and efficacy. EXPERIMENTAL DESIGN DRUP is a multicenter, nonrandomized clinical basket trial treating patients with therapy-refractory cancer with molecularly targeted and immunotherapies outside their registered indications (NCT02925234). Here, all granted waivers were revised, analyzed in terms of safety and efficacy outcome, and comparedwithoutcomes of includedpatientswho didnot receive awaiver. RESULTS Between September 1, 2016, and September 1, 2021, protocol waivers were granted for 82 patients (8%) of 1,019 included patients in DRUP. Most waivers (45%) were granted for general- or drug-related eligibility criteria; other categories were out-of-window testing, treatment, and testing exceptions. Serious adverse event rate was similar between patients who received a waiver (pW) and patients who did not (pNW): 39% vs. 41%, respectively (P = 0.81). The clinical benefit (either objective response or stable disease ≥ 16 weeks) rate of pW was 40% versus 33% in pNW (P = 0.43). CONCLUSIONS Safety and clinical benefit were preserved in patients for whom a waiver was granted. These data support a more personalized approach in assessing eligibility criteria, especially in trials with widely used and approved drugs accruing patients without other treatment options. See related commentary by Waqar and Govindan, p. 3655.
Collapse
Affiliation(s)
| | - Laurien J Zeverijn
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Birgit S Geurts
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Louisa R Hoes
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | | | | | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Filip Y F de Vos
- Department of Medical Oncology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Katrien Grünberg
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Haiko J Bloemendal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Emile E Voest
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
4
|
Bogdan L, Saleh RR, Avery L, Del Rossi S, Yu C, Bedard PL. Clinical Utility of Tumor Next-Generation Sequencing Panel Testing to Inform Treatment Decisions for Patients With Advanced Solid Tumors in a Tertiary Care Center. JCO Precis Oncol 2024; 8:e2400092. [PMID: 38935894 PMCID: PMC11371095 DOI: 10.1200/po.24.00092] [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: 02/06/2024] [Revised: 03/28/2024] [Accepted: 05/02/2024] [Indexed: 06/29/2024] Open
Abstract
PURPOSE There is limited information about the clinical utility of targeted next-generation sequencing (NGS) panel testing to inform decision making for patients with advanced solid tumors. The Ontario-wide Cancer Targeted Nucleic Acid Evaluation (OCTANE) is a prospective study that enrolled more than 4,500 patients with solid tumor for NGS panel testing. We performed a retrospective survey of medical oncologists to evaluate the impact of NGS testing on treatment decisions. METHODS Patients and treating oncologists were identified at the Princess Margaret Cancer Center between 2016 and 2021. Tumor-only sequencing was performed using a gene panel of either 555 or 161 cancer genes. Oncologists were asked to review testing results and complete a survey indicating whether NGS testing affected treatment decisions. The primary outcome of this study was rate of treatment change on the basis of mutation results. Patient, test, and physician factors were evaluated for association with treatment changes using univariate analyses and a mixed-effects model. RESULTS Of the 582 surveys sent, 394 (67.7%) were completed. We found that 188 (47.7%) patients had testing results classified as actionable by the oncologist and 62 (15.7%) patients were matched to treatment, of whom 37 (60%) were enrolled in a clinical trial, 13 (21%) received an approved drug, four (6%) were prescribed off-label therapy, and eight (13%) avoided ineffective treatment. Patient, test, and physician characteristics were not significantly associated with treatment change. There was no difference in overall survival between patients who received matched treatment versus those who did not (P = .55, median survival not reached). CONCLUSION OCTANE testing led to a change in drug treatment in 15.7% of patients, supporting the clinical utility of NGS panel testing for patients with advanced solid tumors.
Collapse
Affiliation(s)
- Lucia Bogdan
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Canada
| | - Ramy R. Saleh
- Department of Medical Oncology, McGill University Health Centre, Montreal, Canada
| | - Lisa Avery
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Samanta Del Rossi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Celeste Yu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Philippe L. Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
5
|
Gimeno-Valiente F, Martín-Arana J, Tébar-Martínez R, Gambardella V, Martínez-Ciarpaglini C, García-Micó B, Martínez-Castedo B, Palomar B, García-Bartolomé M, Seguí V, Huerta M, Moro-Valdezate D, Pla-Martí V, Pérez-Santiago L, Roselló S, Roda D, Cervantes A, Tarazona N. Sequencing paired tumor DNA and white blood cells improves circulating tumor DNA tracking and detects pathogenic germline variants in localized colon cancer. ESMO Open 2023; 8:102051. [PMID: 37951129 PMCID: PMC10774972 DOI: 10.1016/j.esmoop.2023.102051] [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: 07/27/2023] [Accepted: 09/22/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND In the setting of localized colon cancer (CC), circulating tumor DNA (ctDNA) monitoring in plasma has shown potential for detecting minimal residual disease (MRD) and predicting a higher risk of recurrence. With the tumor-only sequencing approach, however, germline variants may be misidentified as somatic variations, precluding the possibility of tracking in up to 11% of patients due to a lack of known somatic mutations. In this study, we assess the potential value of adding white blood cells (WBCs) to tumor tissue sequencing to enhance the accuracy of sequencing results. PATIENTS AND METHODS A total of 148 patients diagnosed with localized CC were prospectively recruited at the Hospital Clínico Universitario in Valencia (Spain). Employing a custom 29-gene panel, sequencing was conducted on tumor tissue, plasma and corresponding WBCs. Droplet digital PCR and amplicon-based NGS were performed on plasma samples post-surgery to track MRD. Oncogenic somatic variants were identified by annotating with COSMIC, OncoKB and an internal repository of pathogenic mutations database. A variant prioritization analysis, mainly characterized by the match of oncogenic mutations with the evidence levels defined in OncoKB, was carried out to select specific targeted therapies. RESULTS Utilizing paired tumor and WBCs sequencing, we identified somatic mutations in all patients (100%) within our cohort, compared to 89% using only tumor tissue. Consequently, the top 10 most frequently mutated genes for plasma monitoring were altered. The sequencing of WBCs identified 9% of patients with pathogenic mutations in the germline, with APC and TP53 being the most frequently mutated genes. Additionally, mutations in genes related to clonal hematopoiesis of indeterminate potential were detected in 27% of the cohort, with TP53, KRAS, and KMT2C being the most frequently altered genes. There were no observed differences in the sensitivity of monitoring MRD using ddPCR or amplicon-based NGS (p = 1). Ultimately, 41% of the patients harbored potentially targetable alterations at diagnosis. CONCLUSION The germline testing method not only enhanced sequencing results and raised the proportion of patients eligible for plasma monitoring, but also uncovered the existence of pathogenic germline variations, thereby aiding in the identification of patients at a higher risk of hereditary cancer syndromes.
Collapse
Affiliation(s)
- F Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - J Martín-Arana
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid
| | - R Tébar-Martínez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - V Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - C Martínez-Ciarpaglini
- CIBERONC, Instituto de Salud Carlos III, Madrid; Department of Pathology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - B García-Micó
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid
| | - B Martínez-Castedo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid
| | - B Palomar
- Department of Pathology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - M García-Bartolomé
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - V Seguí
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - M Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia
| | - D Moro-Valdezate
- Colorectal Surgery Unit, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, Department of Surgery, University of Valencia, Valencia, Spain
| | - V Pla-Martí
- Colorectal Surgery Unit, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, Department of Surgery, University of Valencia, Valencia, Spain
| | - L Pérez-Santiago
- Colorectal Surgery Unit, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, Department of Surgery, University of Valencia, Valencia, Spain
| | - S Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid
| | - D Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid
| | - A Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid.
| | - N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid.
| |
Collapse
|
6
|
Weiss L, Dorman K, Boukovala M, Schwinghammer F, Jordan P, Fey T, Hasselmann K, Subklewe M, Bücklein V, Bargou R, Goebeler M, Sayehli C, Spoerl S, Lüke F, Heudobler D, Claus R, von Luettichau I, Lorenzen S, Lange S, Westphalen CB, von Bergwelt-Baildon M, Heinemann V, Gießen-Jung C. Early clinical trial unit tumor board: a real-world experience in a national cancer network. J Cancer Res Clin Oncol 2023; 149:13383-13390. [PMID: 37490102 PMCID: PMC10587227 DOI: 10.1007/s00432-023-05196-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Early clinical trials are the first step into clinical therapies for new drugs. Within the six Bavarian university-based hospitals (Augsburg, Erlangen, Regensburg, Munich (LMU and TU), Würzburg) we have enrolled a virtual network platform for patient discussion. METHODS The virtual Early Clinical Trial Unit Tumor Board (ECTU Tumor Board) is a secured web-based meeting to evaluate early clinical trial options for patients, where representatives from local ECTUs participate. We retrospectively analyzed patient cases discussed between November 2021 and November 2022. RESULTS From November 2021 to November 2022, a total of 43 patients were discussed in the ECTU Tumor Board. Median age at diagnosis was 44.6 years (range 10-76 years). The median number of previous lines of therapies was 3.7 (range 1-9 therapies) including systemic treatment, surgery, and radiation therapy. A total of 27 different tumor entities were presented and 83.7% (36/43) patients received at least one trial recommendation. In total, 21 different active or shortly recruiting clinical trials were recommended: ten antibody trials, four BiTE (bispecific T cell engager) trials, six CAR (chimeric antigen receptor) T-cell trials, and one chemotherapy trial. Only six trials (28.6%) were recommended on the basis of the previously performed comprehensive genetic profiling (CGP). CONCLUSION The ECTU Tumor Board is a feasible and successful network, highlighting the force of virtual patient discussions for improving patient care as well as trial recruitment in advanced diseases. It can provide further treatment options after local MTB presentation, aiming to close the gap to access clinical trials.
Collapse
Affiliation(s)
- L Weiss
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - K Dorman
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M Boukovala
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - F Schwinghammer
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - P Jordan
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - T Fey
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - K Hasselmann
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - M Subklewe
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - V Bücklein
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - R Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M Goebeler
- Early Clinical Trials Unit, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C Sayehli
- Early Clinical Trials Unit, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Spoerl
- Department of Internal Medicine 5 (Hematology and Clinical Oncology), Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - F Lüke
- Department of Internal Medicine III (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - D Heudobler
- Department of Internal Medicine III (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - R Claus
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - I von Luettichau
- Department of Pediatrics and Children's Cancer Research Center, TUM School of Medicine, Kinderklinik München Schwabing, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Lorenzen
- Department of Medicine II (Gastroenterology), Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - S Lange
- Department of Medicine II (Gastroenterology), Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C B Westphalen
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - M von Bergwelt-Baildon
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - V Heinemann
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - C Gießen-Jung
- Department Medicine III (Hematology and Oncology), LMU University Hospital Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
7
|
Teuwen LA, Roets E, D’Hoore P, Pauwels P, Prenen H. Comprehensive Genomic Profiling and Therapeutic Implications for Patients with Advanced Cancers: The Experience of an Academic Hospital. Diagnostics (Basel) 2023; 13:1619. [PMID: 37175010 PMCID: PMC10177779 DOI: 10.3390/diagnostics13091619] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Next-generation sequencing (NGS) can be used to detect tumor-specific genomic alterations. This retrospective single-center study aims to assess the application of an extensive NGS panel to identify actionable alterations and initiate matched targeted treatment for patients with advanced cancer. We analyzed genomic alterations in solid tumor biopsies from 464 patients with advanced cancer with the Foundation Medicine assay (FoundationOne®CDx). Therapeutic implications were determined using the Memorial Sloan Kettering Precision Oncology Knowledge Base (OncoKB) classification. The FoundationOne®CDx was successfully applied in 464/521 patients (89%). The most common altered genes were TP53 (61%), KRAS (20%), CDKN2A (20%), TERT (16%), and APC (16%). Among the 419 patients with successfully analyzed tumor mutational burden (TMB), 43 patients presented with a high TMB (≥10 mutations/megabase). Out of the 126 patients with an actionable target, 40 patients received matched treatment (32%) of which 17 were within a clinical trial. This study shows that the application of NGS is feasible in an academic center and increases the detection of actionable alterations and identification of patients eligible for targeted treatment or immunotherapy regardless of tumor histology. Strategies such as early referral for NGS, inclusion in clinical (basket) trials, and the development of new targeted drugs are necessary to improve the matched treatment rate.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Evelyne Roets
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Pieter D’Hoore
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Patrick Pauwels
- Department of Pathology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium;
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Hans Prenen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
8
|
Carmagnani Pestana R, Moyers JT, Roszik J, Sen S, Hong DS, Naing A, Herzog CE, Fu S, Piha-Paul SA, Rodon J, Yap TA, Karp DD, Tsimberidou AM, Pant S, Zarzour MA, Ratan R, Ravi V, Benjamin RS, Lazar AJ, Wang WL, Daw N, Gill JB, Harrison DJ, Lewis VO, Roland CL, Patel SR, Livingston JA, Somaiah N, Ludwig JA, Conley AP, Hamerschlak N, Gorlick R, Meric-Bernstam F, Subbiah V. Impact of Biomarker-Matched Therapies on Outcomes in Patients with Sarcoma Enrolled in Early-Phase Clinical Trials (SAMBA 101). Clin Cancer Res 2023; 29:1708-1718. [PMID: 37058010 PMCID: PMC10150251 DOI: 10.1158/1078-0432.ccr-22-3629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/30/2022] [Accepted: 02/24/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE Developing new therapeutics for any of the more than 100 sarcoma subtypes presents a challenge. After progression from standard therapies, patients with sarcoma may be referred for enrollment in early-phase trials. This study aimed to investigate whether enrollment in biomarker-matched early-phase clinical trials leads to better outcomes for patients with advanced sarcoma. EXPERIMENTAL DESIGN In this retrospective analysis, investigational treatment characteristics and longitudinal survival outcomes were analyzed in patients with biopsy-confirmed sarcoma enrolled in early-phase trials at MD Anderson Cancer Center from May 2006 to July 2021. RESULTS Five hundred eighty-seven patients were included [405 soft tissue, 122 bone, 60 gastrointestinal stromal tumor (GIST); median of three prior lines of therapy]. Most common subtypes were leiomyosarcoma (17.2%), liposarcoma (14.0%), and GIST (10.2%). Molecular testing was available for 511 patients (87.1%); 221 patients (37.6%) were treated in matched trials. Overall response rate was 13.1% matched compared with 4.9% in unmatched (P < 0.001); the clinical benefit rate at 6 months was 43.9% vs. 19.9% (P < 0.001). Progression-free survival was longer for patients in matched trials (median, 5.5 vs. 2.4 months; P < 0.001), and overall survival was also superior for patients in matched trials (median, 21.5 vs. 12.3 months; P < 0.001). The benefit of enrollment in matched trials was maintained when patients with GIST were excluded from the analysis. CONCLUSIONS Enrollment in biomarker-matched early-phase trials is associated with improved outcomes in heavily pretreated patients with metastatic sarcoma. Molecular testing of tumors from patients with advanced sarcoma and enrollment in matched trials is a reasonable therapeutic strategy.
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Justin T. Moyers
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Medicine, Division of Hematology and Oncology, The University of California, Irvine, Orange, California
| | - Jason Roszik
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiraj Sen
- Sarah Cannon Research Institute, Nashville, Tennessee
| | - David S. Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia E. Herzog
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina A. Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy A. Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel D. Karp
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria A. Zarzour
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S. Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J. Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Najat Daw
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan B. Gill
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas J. Harrison
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Valerae O. Lewis
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shreyaskumar R. Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J. Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph A. Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P. Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Richard Gorlick
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Papaccio F, Cabeza-Segura M, Garcia-Micò B, Tarazona N, Roda D, Castillo J, Cervantes A. Will Organoids Fill the Gap towards Functional Precision Medicine? J Pers Med 2022; 12:1939. [PMID: 36422115 PMCID: PMC9695811 DOI: 10.3390/jpm12111939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 08/13/2023] Open
Abstract
Precision medicine approaches for solid tumors are mainly based on genomics. Its employment in clinical trials has led to somewhat underwhelming results, except for single responses. Moreover, several factors can influence the response, such as gene and protein expression, the coexistence of different genomic alterations or post-transcriptional/translational modifications, the impact of tumor microenvironment, etc., therefore making it insufficient to employ a genomics-only approach to predict response. Recently, the implementation of patient-derived organoids has shed light on the possibility to use them to predict patient response to drug treatment. This could offer for the first time the possibility to move precision medicine to a functional environment.
Collapse
Affiliation(s)
- Federica Papaccio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Manuel Cabeza-Segura
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
| | - Blanca Garcia-Micò
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
| | - Noelia Tarazona
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
- Centro de Investigacion Biomedica en Red (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Desamparados Roda
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
- Centro de Investigacion Biomedica en Red (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josefa Castillo
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
- Centro de Investigacion Biomedica en Red (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
| | - Andres Cervantes
- Biomedical Research Institute INCLIVA, Hospital Clínico Universitario, Department Medical Oncology, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
10
|
An Analysis of Transcriptomic Burden Identifies Biological Progression Roadmaps for Hematological Malignancies and Solid Tumors. Biomedicines 2022; 10:biomedicines10112720. [DOI: 10.3390/biomedicines10112720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Biological paths of tumor progression are difficult to predict without time-series data. Using median shift and abacus transformation in the analysis of RNA sequencing data sets, natural patient stratifications were found based on their transcriptomic burden (TcB). Using gene-behavior analysis, TcB groups were evaluated further to discover biological courses of tumor progression. We found that solid tumors and hematological malignancies (n = 4179) share conserved biological patterns, and biological network complexity decreases at increasing TcB levels. An analysis of gene expression datasets including pediatric leukemia patients revealed TcB patterns with biological directionality and survival implications. A prospective interventional study with PI3K targeted therapy in canine lymphomas proved that directional biological responses are dynamic. To conclude, TcB-enriched biological mechanisms detected the existence of biological trajectories within tumors. Using this prognostic informative novel informatics method, which can be applied to tumor transcriptomes and progressive diseases inspires the design of progression-specific therapeutic approaches.
Collapse
|
11
|
Abstract
SummaryThe biology of non-small cell lung cancer (NSCLC) is driven by a complex mutational landscape, and the detection of driver molecular alterations by next-generation sequencing is key for identification of druggable alterations. Thus, broad molecular profiling displays a standard-of-care approach particularly in patients with advanced adenocarcinoma at the time of the initial diagnosis, but also at the time of acquired resistance to tyrosine kinase inhibitors, guiding further treatment choices. Sequencing of plasma-circulating tumor DNA is of increasing importance in NSCLC diagnostics due to the easy accessibility, representing an optimal tool for longitudinal monitoring of the disease course.
Collapse
|
12
|
Takeda H, Imoto K, Umemoto K, Doi A, Arai H, Horie Y, Mizukami T, Oguri T, Ogura T, Izawa N, Yamamoto H, Yamano Y, Sunakawa Y. Clinical Utility of Genomic Profiling Tests in Patients with Advanced Gastrointestinal Cancers. Target Oncol 2022; 17:177-185. [PMID: 35366174 DOI: 10.1007/s11523-022-00871-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Comprehensive analyses of cancer-related genomic alterations are expected to lead to increased availability of targeted therapies. However, in patients with gastrointestinal (GI) cancers, the utility of genomic profiling is unclear because of common non-druggable alterations and rapid disease progression that prevent a sufficient time period to seek targets. OBJECTIVE The aim of this study was to determine the utility of genomic profiling tests in patients with GI cancers. METHODS The subjects of this retrospective study were patients with GI cancers and patients with non-GI cancers who underwent tissue-based genomic profiling at a single institution from April 2017 to October 2020. The profile of gene alterations, frequency of tumor mutational burden-high (≥ 10 Muts/Mb), and accessibility of recommended molecular targeted therapy were compared between patients with GI cancers and patients with non-GI cancers. RESULTS In all, 133 patients with GI cancers and 63 patients with non-GI cancers were included. The genomic profiles of GI cancers showed the highest frequencies of TP53, KRAS, and APC mutations and a significantly lower frequency of PIK3CA mutations than those of non-GI cancers. Tumor mutational burden-high was significantly less prevalent in GI cancers (4% vs 20%, p = 0.008). Twenty-nine patients with GI cancers (40%) and 35 patients with non-GI cancers (56%) were recommended for targeted therapies based on the findings. Among them, seven patients each with GI cancers and non-GI cancers received the recommended therapy on their genomic findings, which showed similar treatment accessibility between the GI and non-GI cancer groups (10% vs 11%, p = 0.791). HER2-targeted and BRAF-targeted therapies were the primary treatments administered to patients with GI cancers. CONCLUSIONS Although their genomic profiles revealed fewer druggable sites, patients with GI cancers accessed targeted therapies similarly to patients with non-GI cancers. The utility of genomic profile testing in patients with GI cancers was highlighted to determine if patients can receive specific treatments, such as HER2-targeted and BRAF-targeted therapies.
Collapse
Affiliation(s)
- Hiroyuki Takeda
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kiyomi Imoto
- Center for Genomic Medicine, St. Marianna University Hospital, Kawasaki, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ayako Doi
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Arai
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshiki Horie
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takuro Mizukami
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomoyo Oguri
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takashi Ogura
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Izawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Yamamoto
- Department of Bioinformatics, St. Marianna University Graduate School of Medicine, Kawasaki, Japan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshihisa Yamano
- Division of Neurology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan. .,Center for Genomic Medicine, St. Marianna University Hospital, Kawasaki, Japan.
| |
Collapse
|
13
|
Burton KA, Mahen E, Konnick EQ, Blau S, Dorschner MO, Ramirez AB, Schmechel SC, Song C, Parulkar R, Parker S, Senecal FM, Pritchard CC, Mecham BH, Szeto C, Spilman P, Zhu J, Gadi VK, Ronen R, Stilwell J, Kaldjian E, Dutkowski J, Benz SC, Rabizadeh S, Soon-Shiong P, Blau CA. Safety, Feasibility, and Merits of Longitudinal Molecular Testing of Multiple Metastatic Sites to Inform mTNBC Patient Treatment in the Intensive Trial of Omics in Cancer. JCO Precis Oncol 2022; 6:e2100280. [PMID: 35294224 PMCID: PMC8939922 DOI: 10.1200/po.21.00280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Patients with metastatic triple-negative breast cancer (mTNBC) have poor outcomes. The Intensive Trial of Omics in Cancer (ITOMIC) sought to determine the feasibility and potential efficacy of informing treatment decisions through multiple biopsies of mTNBC deposits longitudinally over time, accompanied by analysis using a distributed network of experts. In the Intensive Trial of Omics in Cancer (ITOMIC), the feasibility and potential efficacy of informing treatment decisions through omics analysis of multiple biopsies of mTNBC deposits over time was assessed. An ITOMIC Tumor Board (ITB) that comprised experts discussed tumor profile findings and made treatment recommendations to each subject's physician. Study-directed omics analysis revealed that of the 31 enrolled subjects, two were found to have lung cancer, one a carcinoma of unknown primary site that and tumor samples from five subjects showed some receptor-positivity. Several subjects survived well beyond what would be expected for this patient group, supporting the merits of further investigation of this approach.![]()
Collapse
Affiliation(s)
- Kimberly A Burton
- Department of Medicine, University of Washington, Seattle, WA.,Center for Cancer Innovation, University of Washington, Seattle, WA.,Northwest Medical Specialties, Puyallup and Tacoma, WA.,South Sound CARE Foundation, Seattle, WA
| | - Elisabeth Mahen
- Center for Cancer Innovation, University of Washington, Seattle, WA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA.,Department of Medicine/Hematology, University of Washington, Seattle, WA
| | | | - Sibel Blau
- Center for Cancer Innovation, University of Washington, Seattle, WA.,Northwest Medical Specialties, Puyallup and Tacoma, WA
| | - Michael O Dorschner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA.,Center for Precision Diagnostics, University of Washington, Seattle, WA
| | | | - Stephen C Schmechel
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Chaozhong Song
- Center for Cancer Innovation, University of Washington, Seattle, WA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA.,Department of Medicine/Hematology, University of Washington, Seattle, WA
| | | | - Stephanie Parker
- Northwest Medical Specialties, Puyallup and Tacoma, WA.,South Sound CARE Foundation, Seattle, WA
| | - Francis Mark Senecal
- Northwest Medical Specialties, Puyallup and Tacoma, WA.,South Sound CARE Foundation, Seattle, WA
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | | | | | | | - Jingchun Zhu
- Computational Genomics Lab, University of California at Santa Cruz, Santa Cruz, CA
| | - Vijayakrishna K Gadi
- Department of Medicine, University of Illinois, Chicago, IL.,Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | | | | | | | - C Anthony Blau
- Center for Cancer Innovation, University of Washington, Seattle, WA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA.,Department of Medicine/Hematology, University of Washington, Seattle, WA.,All4Cure Inc, Seattle, WA
| |
Collapse
|
14
|
[Molecular diagnostics and molecular tumor board in uro-oncology : Precision medicine using the example of metastatic castration-resistant prostate cancer]. Urologe A 2022; 61:311-322. [PMID: 35157098 DOI: 10.1007/s00120-022-01784-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 10/19/2022]
Abstract
Novel approaches to molecular tumor profiling evaluate DNA, RNA and protein alterations to create a detailed molecular map that enables precise and personalized treatment decisions. As the field of molecular profiling is constantly evolving, the training and networking of doctors is of decisive importance. Through the establishment of precision medicine with precision oncological consultations supported by interdisciplinary molecular tumor boards, many patients with difficult to treat tumor diseases can be advised and treated. Many pathophysiological relationships in progressive tumors can be elucidated resulting in new therapeutic options for the profiled patients; however, understanding the complex mutational profiles remains a very demanding task that requires a suitably trained and committed team that should be in close contact with the scientific advancements in precision oncology.
Collapse
|
15
|
Napolitano S, Caputo V, Ventriglia A, Martini G, Della Corte CM, De Falco V, Ferretti S, Martinelli E, Morgillo F, Ciardiello D, De Vita F, Orditura M, Fasano M, Ciardiello F, Troiani T. OUP accepted manuscript. Oncologist 2022; 27:e633-e641. [PMID: 35604409 PMCID: PMC9355826 DOI: 10.1093/oncolo/oyac071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
CoronaVirus disease-2019 has changed the delivery of health care worldwide and the pandemic has challenged oncologists to reorganize cancer care. Recently, progress has been made in the field of precision medicine to provide to patients with cancer the best therapeutic choice for their individual needs. In this context, the Foundation Medicine (FMI)-Liquid@Home project has emerged as a key weapon to deal with the new pandemic situation. FoundationOne Liquid Assay (F1L) is a next-generation sequences-based liquid biopsy service, able to detect 324 molecular alterations and genomic signatures, from May 2020 available at patients’ home (FMI-Liquid@Home). We analyzed time and costs saving for patients with cancer, their caregivers and National Healthcare System (NHS) with FMI-Liquid@Home versus F1L performed at our Department. Different variables have been evaluated. Between May 2020 and August 2021, 218 FMI-Liquid@Home were performed for patients with cancer in Italy. Among these, our Department performed 153 FMI-Liquid@Home with the success rate of 98% (vs. 95% for F1L in the hospital). Time saving for patients and their caregivers was 494.86 and 427.36 hours, respectively, and costs saving was 13 548.70€. Moreover, for working people these savings were 1084.71 hours and 31 239.65€, respectively. In addition, the total gain for the hospital was 163.5 hours and 6785€, whereas for NHS was 1084.71 hours and 51 573.60€, respectively. FMI-Liquid@Home service appears to be useful and convenient allowing time and costs saving for patients, caregivers, and NHS. Born during the COVID-19 pandemic, it could be integrated in oncological daily routine in the future. Therefore, additional studies are needed to better understand the overall gain and how to integrate this service in different countries.
Collapse
Affiliation(s)
| | | | - Anna Ventriglia
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Giulia Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Carminia Maria Della Corte
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Vincenzo De Falco
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | | | - Erika Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Floriana Morgillo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Davide Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
- Oncology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Foggia, Italy
| | - Ferdinando De Vita
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Michele Orditura
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Morena Fasano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Teresa Troiani
- Corresponding author: Teresa Troiani, Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, Via S. Pansini 5, 80131, Napoli, Italy. Tel: +39 0815666729;
| |
Collapse
|
16
|
Russo A, Incorvaia L, Capoluongo E, Tagliaferri P, Galvano A, Del Re M, Malapelle U, Chiari R, Conte P, Danesi R, Fassan M, Ferrara R, Genuardi M, Ghiorzo P, Gori S, Guadagni F, Marchetti A, Marchetti P, Midiri M, Normanno N, Passiglia F, Pinto C, Silvestris N, Tallini G, Vatrano S, Vincenzi B, Cinieri S, Beretta G. The challenge of the Molecular Tumor Board empowerment in clinical oncology practice: A Position Paper on behalf of the AIOM- SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies. Crit Rev Oncol Hematol 2021; 169:103567. [PMID: 34896250 DOI: 10.1016/j.critrevonc.2021.103567] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022] Open
Abstract
The development of innovative technologies and the advances in the genetics and genomics, have offered new opportunities for personalized treatment in oncology. Although the selection of the patient based on the molecular characteristics of the neoplasm has the potential to revolutionize the therapeutic scenario of oncology, this approach is extremely challenging. The access, homogeneity, and economic sustainability of the required genomic tests should be warranted in the clinical practice, as well as the specific scientific and clinical expertise for the choice of medical therapies. All these elements make essential the collaboration of different specialists within the Molecular Tumor Boards (MTBs). In this position paper, based on experts' opinion, the AIOM-SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies critically discuss the available molecular profiling technologies, the proposed criteria for the selection of patients candidate for evaluation by the MTB, the criteria for the selection and analysis of biological samples, and the regulatory and pharmaco-economic issues.
Collapse
Affiliation(s)
- Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy.
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Ettore Capoluongo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy; CEINGE, Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80131, Naples, Italy
| | - Pierosandro Tagliaferri
- Medical and Translational Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80138, Naples, Italy
| | - Rita Chiari
- Medical Oncology, AULSS 6 Euganea, South Padova Hospital, Monselice, PD, Italy
| | - Pierfranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Medical Oncology 2, Istituto Oncologico Veneto - IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy; Veneto Institute of Oncology, IOV - IRCCS, Padua, Italy
| | - Roberto Ferrara
- Department of Medical Oncology, Thoracic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Maurizio Genuardi
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy; UOC Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Paola Ghiorzo
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties, University of Genoa, 16132, Genoa, Italy
| | - Stefania Gori
- Department of Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Fiorella Guadagni
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, University-Foundation, CeSI Biotech Chieti, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Massimo Midiri
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, Turin, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Centre, IRCCS-AUSL di Reggio Emilia, Reggio Emilia, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology, Department of Internal Medicine and Oncology (DIMO), University of Bari, Bari, Italy
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138, Bologna, Italy
| | - Simona Vatrano
- Department of Pathology, Cannizzaro Hospital, Catania, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University, 00128, Rome, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | | |
Collapse
|