1
|
Moretto R, Germani MM, Carullo M, Conca V, Minelli A, Giordano M, Bruno R, Rossini D, Gusmaroli E, De Grandis MC, Antoniotti C, Salvatore L, Passardi A, Tamberi S, Scartozzi M, Pietrantonio F, Lonardi S, Ugolini C, Masi G, Cremolini C. Exploring the Prognostic and Predictive Impact of Genomic Loss of Heterozygosity and Homologous Recombination Deficiency Alterations in Patients With Metastatic Colorectal Cancer. JCO Precis Oncol 2025; 9:e2400567. [PMID: 40249885 DOI: 10.1200/po-24-00567] [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: 08/08/2024] [Revised: 12/20/2024] [Accepted: 03/10/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Genomic loss-of-heterozygosity (gLOH) consists in the loss of chromosomal regions and is associated with homologous recombination repair (HRR) system deficiency. We explored the role of gLOH and HRR-related gene alterations in metastatic colorectal cancer (mCRC). METHODS FoundationOne CDx assay was used to determine the percentage of gLOH and the presence of alterations in 27 HRR-related genes in archival chemo-naïve tumor tissues of patients with mCRC treated with first-line oxaliplatin- or irinotecan-based doublets and triplet ± anti-PD-L1. RESULTS Overall, 243 samples were analyzed. None of the nine deficient mismatch repair/microsatellite instability high tumors were gLOH-high, while 16 (7%) of 234 proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors were gLOH-high. In the pMMR/MSS population, six (3%) and 18 (8%) had at least a biallelic or monoallelic HRR-related gene alteration, respectively. Among patients receiving FOLFOXIRI alone (n = 68) or with an anti-PD-L1 (N = 90), higher benefit from the addition of the immune checkpoint inhibitor (ICI) was observed in the gLOH-high subgroup (n = 12), in terms of both progression-free survival (PFS; Pint = .02) and overall survival (OS; Pint = .03). No differences in PFS or OS were reported between patients treated with first-line oxaliplatin- (n = 40) versus irinotecan-based doublets (n = 25) or with the triplet FOLFOXIRI (n = 68) versus doublets (n = 65), according to the gLOH status. Among patients not receiving an anti-PD-L1, longer PFS was observed in the gLOH-low group (n = 138) versus the gLOH-high (n = 6) group (5.1 v 12.1 months; hazard ratio, 8.73 [95% CI, 3.64 to 20.9]; P < .001), and this was confirmed in the multivariate analysis (P < .001). No prognostic impact of monoallelic or biallelic HRR-related gene alterations was shown. CONCLUSION In pMMR/MSS mCRC, gLOH-high was associated with worse prognosis and higher benefit from the addition of anti-PD-L1 agents to chemotherapy. If confirmed in larger series, these results may inform the design of clinical trials.
Collapse
Affiliation(s)
- Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Martina Carullo
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Veronica Conca
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Minelli
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Clinical Oncology Unit, San Paolo Hospital, Civitavecchia, Italy
| | - Mirella Giordano
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rossella Bruno
- Unit of Pathological Anatomy, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Daniele Rossini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Eleonora Gusmaroli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Caterina De Grandis
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lisa Salvatore
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Stefano Tamberi
- Oncology Unit, Ravenna Hospital, AUSL Romagna, Ravenna, Italy
| | - Mario Scartozzi
- Medical Oncology, University of Cagliari, Via Università, Cagliari, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Clara Ugolini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
2
|
Marchetti C, Ergasti R, Capomacchia FM, Giannarelli D, Mastrantoni L, Pepe F, Apostol AI, Sassu CM, Nero C, Piermattei A, Zannoni GF, Troncone G, Colomban O, Russo G, Carrot A, Malapelle U, You B, Lorusso D, Scambia G, Fagotti A. Integrating clinical-molecular data to predict PARP inhibitors efficacy in advanced ovarian cancer patients after interval cytoreductive surgery. Gynecol Oncol 2025; 195:16-25. [PMID: 40048982 DOI: 10.1016/j.ygyno.2025.02.016] [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: 09/11/2024] [Revised: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVE Selecting the maintenance strategy for advanced tubo-ovarian high-grade serous carcinoma (HGSC) is challenging. This study evaluates the correlation among chemotherapy response score (CRS), homologous recombination deficiency (HRD) status, and KELIM score; identifies predictors of Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) efficacy and stratifies recurrence risk in PARPi-treated population. METHODS Median Progression-free Survival (mPFS) and hazard ratios (HRs) were retrospectively calculated in HGSC patients after neoadjuvant chemotherapy (3/4 cycles), interval cytoreductive surgery, and adjuvant treatment. Variables included HRD status, disease stage, KELIM, radiological response, residual tumor, and CRS at surgery. A risk-stratification model predicting PARPi efficacy was developed. RESULTS Among overall population (N = 373), 66.9 % of CRS3 patients reached favorable KELIM, 17.3 % had complete radiological response, and 97.8 % achieved complete surgery, with higher frequencies than CRS1/2 (p < 0.001). Univariate analysis of PFS on PARPi (N = 210) showed favorable covariates: CRS3 (HR 2.37, 95 % CI 1.39-4.04 and HR 1.59, 95 % CI 1.03-2.47 vs CRS1 and CRS2), BRCA mutation (HR 3.41 95 % CI 2.15-5.39 and HR 2.00 95 % CI 1.13-3.56 vs BRCAwt-HRDneg and -HRDpos) and continuum KELIM (HR 0.66, 95 % CI 0.45-0.96). At multivariate, CRS3 and BRCA mutation were confirmed significant. Combining HRD status, CRS, and KELIM four prognostic groups with different PARPi efficacy were identified (mPFS 38 vs 26 vs 18 vs 6 months for Low, Intermediate, High-Intermediate, and High-risk groups). CONCLUSIONS CRS is a prognostic factor in PARPi-treated population as a PARPi efficacy surrogate. Integrating HRD status, CRS, and KELIM allows patients risk stratification and tailored maintenance. These results should be considered hypothesis-generating.
Collapse
Affiliation(s)
- Claudia Marchetti
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Raffaella Ergasti
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Filippo Maria Capomacchia
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics - GSTeP, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Mastrantoni
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Adriana Ionelia Apostol
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carolina Maria Sassu
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Camilla Nero
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessia Piermattei
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Gian Franco Zannoni
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Olivier Colomban
- Université Lyon, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon-Sud, EA 3738 CICLY, Lyon, France; Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL EPSISLYON, Lyon, France
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Aurore Carrot
- Université Lyon, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon-Sud, EA 3738 CICLY, Lyon, France
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Benoit You
- Université Lyon, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon-Sud, EA 3738 CICLY, Lyon, France; Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL EPSISLYON, Lyon, France
| | - Domenica Lorusso
- Unità di Ginecologia oncologica medica, Humanitas University, Milan, Italy
| | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Fagotti
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
3
|
Witz A, Dardare J, Betz M, Michel C, Husson M, Gilson P, Merlin JL, Harlé A. Homologous recombination deficiency (HRD) testing landscape: clinical applications and technical validation for routine diagnostics. Biomark Res 2025; 13:31. [PMID: 39985088 PMCID: PMC11846297 DOI: 10.1186/s40364-025-00740-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
The use of poly(ADP-ribose) polymerase inhibitors (PARPi) revolutionized the treatment of BRCA-mutated cancers. Identifying patients exhibiting homologous recombination deficiency (HRD) has been proved useful to predict PARPi efficacy. However, obtaining HRD status remains an arduous task due to its evolution over the time. This causes HRD status to become obsolete when obtained from genomic scars, rendering PARPi ineffective for these patients. Only two HRD tests are currently FDA-approved, both based on genomic scars detection and BRCA mutations testing. Nevertheless, new technologies for obtaining an increasingly reliable HRD status continue to evolve. Application of these tests in clinical practice is an additional challenge due to the need for lower costs and shorter time to results delay.In this review, we describe the currently available methods for HRD testing, including the methodologies and corresponding tests for assessing HRD status, and discuss the clinical routine application of these tests and their technical validation.
Collapse
Affiliation(s)
- Andréa Witz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France.
| | - Julie Dardare
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Margaux Betz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Cassandra Michel
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Marie Husson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Pauline Gilson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Jean-Louis Merlin
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Alexandre Harlé
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
4
|
Banerjee S, Eskander RN, Bailey T, Ambler W, Volpe S, Özgören O, Grabowski JP, Valabrega G. Physician knowledge, use, and perceptions of genetic biomarker testing for the management of patients with newly diagnosed advanced ovarian cancer: an international physician survey. Future Oncol 2025; 21:437-445. [PMID: 39815643 PMCID: PMC11812359 DOI: 10.1080/14796694.2025.2449782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025] Open
Abstract
AIMS To explore physician-reported knowledge, use, and perceptions of genetic testing for advanced ovarian cancer management. MATERIALS & METHODS Gynecology/oncology specialists (n = 390) in the US, Europe, Canada, Japan, and Australia completed an online survey spanning March 2021 to April 2022. RESULTS Physician-reported breast cancer gene mutation (BRCAm) testing rates increased over the 2 years before the survey; most patients underwent testing in the preceding 6 months. Homologous recombination deficiency (HRD) genomic instability testing rates and physicians' confidence interpreting results remained relatively low. Genetic testing was driven by the associated treatment implications of the findings. Poor performance status, inadequate tissue, and patients' willingness to undergo testing were reported barriers to testing. CONCLUSIONS Findings indicate that there is a need to improve both access to and information about HRD testing.
Collapse
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Ramez N. Eskander
- San Diego Moores Cancer Center, University of California, San Diego, CA, USA
| | | | | | - Stephanie Volpe
- Global Medical Affairs, Gynecologic Cancers, Oncology R&D, AstraZeneca Pharmaceuticals LP, Cambridge, UK
| | - Ozan Özgören
- Global Medical Affairs, Gynecologic Cancers, Oncology R&D, AstraZeneca Pharmaceuticals LP, Cambridge, UK
| | - Jacek P. Grabowski
- Department of Gynecology, Campus Virchow-Klinikum, Charité Medical University Berlin, Berlin, Germany
| | - Giorgio Valabrega
- Department of Oncology, University of Turin, Division of Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| |
Collapse
|
5
|
Quesada S, Penault-Llorca F, Matias-Guiu X, Banerjee S, Barberis M, Coleman RL, Colombo N, DeFazio A, McNeish IA, Nogueira-Rodrigues A, Oaknin A, Pignata S, Pujade-Lauraine É, Rouleau É, Ryška A, Van Der Merwe N, Van Gorp T, Vergote I, Weichert W, Wu X, Ray-Coquard I, Pujol P. Homologous recombination deficiency in ovarian cancer: Global expert consensus on testing and a comparison of companion diagnostics. Eur J Cancer 2025; 215:115169. [PMID: 39693891 DOI: 10.1016/j.ejca.2024.115169] [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: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Poly (ADP ribose) polymerase inhibitors (PARPis) are a treatment option for patients with advanced high-grade serous or endometrioid ovarian carcinoma (OC). Recent guidelines have clarified how homologous recombination deficiency (HRD) may influence treatment decision-making in this setting. As a result, numerous companion diagnostic assays (CDx) have been developed to identify HRD. However, the optimal HRD testing strategy is an area of debate. Moreover, recently published clinical and translational data may impact how HRD status may be used to identify patients likely to benefit from PARPi use. We aimed to extensively compare available HRD CDx and establish a worldwide expert consensus on HRD testing in primary and recurrent OC. METHODS A group of 99 global experts from 31 different countries was formed. Using a modified Delphi process, the experts aimed to establish consensus statements based on a systematic literature search and CDx information sought from investigators, companies and/or publications. RESULTS Technical information, including analytical and clinical validation, were obtained from 14 of 15 available HRD CDx (7 academic; 7 commercial). Consensus was reached on 36 statements encompassing the following topics: 1) the predictive impact of HRD status on PARPi use in primary and recurrent OC; 2) analytical and clinical validation requirements of HRD CDx; 3) resource-stratified HRD testing; and 4) how future CDx may include additional approaches to help address unmet testing needs. CONCLUSION This manuscript provides detailed information on currently available HRD CDx and up-to-date guidance from global experts on HRD testing in patients with primary and recurrent OC.
Collapse
Affiliation(s)
- Stanislas Quesada
- Department of Medical Oncology, Institut régional du Cancer de Montpellier (ICM), Montpellier, France; Department of Cancer Genetics, University Hospital of Montpellier, Montpellier, France; Groupe d'Investigateurs Nationaux pour l'Etude des cancers de l'ovaire et du sein (GINECO), Paris, France; Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France
| | - Frédérique Penault-Llorca
- Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France; Department of Biology and Pathology, Centre de Lutte Contre le Cancer Jean Perrin, Imagerie Moléculaire et Stratégies Théranostiques, Université Clermont Auvergne, UMR 1240 INSERM-UCA, Clermont-Ferrand, France; Cours St Paul, Saint Paul, Réunion, France
| | - Xavier Matias-Guiu
- Department of Pathology, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, Lleida, Spain; Department of Pathology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain; European Society of Pathology (ESP), Belgium
| | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Massimo Barberis
- Division of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Nicoletta Colombo
- Gynecologic Oncology Program, European Institute of Oncology IRCCS, Milan, Italy; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia; The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Iain A McNeish
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Angélica Nogueira-Rodrigues
- Federal University MG, Brazilian Group of Gynecologic Oncology (EVA), Latin American Cooperative Oncology Group (LACOG), Oncoclínicas, DOM Oncologia, Brazil
| | - Ana Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori di Napoli, IRCCS Fondazione Pascale, Napoli, Italy
| | - Éric Pujade-Lauraine
- Association de Recherche Cancers Gynécologiques - Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'ovaire et du Sein (ARCAGY-GINECO), Paris, France
| | - Étienne Rouleau
- Coordinator of Gen&Tiss GFCO, Université Paris-Saclay, Gustave-Roussy Cancer Campus, Inserm U981, Villejuif, France; Cancer Genetics Laboratory, Medical Biology and Pathology Department, Gustave-Roussy Cancer Campus, Villejuif, France
| | - Aleš Ryška
- European Society of Pathology (ESP), Belgium; The Fingerland Department of Pathology, Faculty of Medicine, Charles University and University Hospital, Hradec Kralove, Czech Republic
| | - Nerina Van Der Merwe
- Division of Human Genetics, National Health Laboratory Service, Universitas Hospital, Bloemfontein, South Africa; Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Toon Van Gorp
- Division of Gynaecological Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium; Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium
| | - Ignace Vergote
- Division of Gynaecological Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium; Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium
| | - Wilko Weichert
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Isabelle Ray-Coquard
- Groupe d'Investigateurs Nationaux pour l'Etude des cancers de l'ovaire et du sein (GINECO), Paris, France; Medical Oncology, Centre Léon Bérard and Université Claude Bernard Lyon, Lyon, France
| | - Pascal Pujol
- Department of Medical Oncology, Institut régional du Cancer de Montpellier (ICM), Montpellier, France; Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France; Center for Ecological and Evolutionary Cancer Research (CREEC), Montpellier University, Montpellier, France.
| |
Collapse
|
6
|
Kechin A, Koryukov M, Mikheeva R, Filipenko M. Homologous recombination deficiency (HRD) diagnostics: underlying mechanisms and new perspectives. Cancer Metastasis Rev 2024; 44:19. [PMID: 39724448 DOI: 10.1007/s10555-024-10238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Homologous recombination deficiency (HRD) is considered a universal and effective sign of a tumor's sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. HRD diagnostics have undergone several stages of transformations: from detection of point mutations in HR-related genes and large regions with loss of heterozygosity detected using single-nucleotide polymorphism arrays to whole-genome signatures of single-nucleotide variants, large genomic rearrangements (LGRs), and copy number alterations. All these methods have their own advantages and limitations. HRD tests, based on signatures of LGRs and copy number alterations, show in hindsight that some progenitor cells have possessed HRD status but not the current state of the genome. The aim of this review was to compare different methods of HRD detection and mechanisms of formation of HRD-specific LGRs. In the last several years, new data appeared implying a crucial role of proteins BRCA1 and BRCA2 in the resolution of stalled replication forks that may be associated with at least some of LGRs observed in HRD-positive tumors. Reviewing current knowledge on these mechanisms, distributions of different LGR types, and limitations of sequencing technologies and algorithms of data analysis, we offer some new perspectives on HRD diagnostics. We hope that this review will help to accelerate the development of new diagnostic approaches in this important field of molecular oncology.
Collapse
Affiliation(s)
- Andrey Kechin
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia.
- Novosibirsk State University, Novosibirsk, 630090, Russia.
| | - Maksim Koryukov
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
- Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Regina Mikheeva
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
- Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Maksim Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| |
Collapse
|
7
|
Tonti N, Golia D’Augè T, Cuccu I, De Angelis E, D’Oria O, Perniola G, Laganà AS, Etrusco A, Ferrari F, Saponara S, Di Donato V, Bogani G, Giannini A. The Role of Tumor Biomarkers in Tailoring the Approach to Advanced Ovarian Cancer. Int J Mol Sci 2024; 25:11239. [PMID: 39457020 PMCID: PMC11508316 DOI: 10.3390/ijms252011239] [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: 09/21/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Growing evidence has demonstrated the role of mutations of tumor biomarkers in diagnosing and treating epithelial ovarian cancer. This review aims to analyze recent literature on the correlation between tumor biomarkers and chemotherapy in nonmucinous ovarian cancer, providing suggestions for personalized treatment approaches. An extensive literature search was conducted to identify relevant studies and trials. BRCA1/2 mutations are central in homologous recombination repair deficiency (HRD) in ovarian cancer, but several other genetic mutations also contribute to varying cancer risks. While the role of MMR testing in ovarian cancer is debated, it is more commonly linked to non-serous ovarian cancer, often associated with Lynch syndrome. A significant proportion of ovarian cancer patients have HRD, affecting treatment decisions in both first-line (especially in advanced stages) and second-line therapy due to HRD's connection with platinum-based therapy and PARP inhibitors' response. However, validated genetic tests to identify HRD have not yet been universally implemented. There is no definitive therapeutic algorithm for advanced ovarian cancer, despite ongoing efforts and multiple proposed tools. Future research should focus on expanding the utility of biomarkers, reducing resistance, and increasing the actionable biomarker pool.
Collapse
Affiliation(s)
- Noemi Tonti
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Tullio Golia D’Augè
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Ilaria Cuccu
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Emanuele De Angelis
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Ottavia D’Oria
- Obstetrics and Gynecological Unit, Department of Woman’s and Child’s Health, San Camillo-Forlanini Hospital, 00152 Rome, Italy;
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Antonio Simone Laganà
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.S.L.); (A.E.)
| | - Andrea Etrusco
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.S.L.); (A.E.)
| | - Federico Ferrari
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Stefania Saponara
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy;
| | - Violante Di Donato
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy; (N.T.); (I.C.); (E.D.A.); (G.P.); (V.D.D.)
| | - Giorgio Bogani
- Gynecological Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Andrea Giannini
- Unit of Gynecology, Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| |
Collapse
|
8
|
Roma C, Esposito Abate R, Sacco A, Califano D, Arenare L, Bergantino F, Pisano C, Cecere SC, Scambia G, Lorusso D, Artioli G, Tasca G, Spina A, Russo D, Gadducci A, De Angelis C, Bologna A, Marchini S, Capoluongo ED, Perrone F, Pignata S, Normanno N. Harmonization of homologous recombination deficiency testing in ovarian cancer: Results from the MITO16A/MaNGO-OV2 trial. Eur J Cancer 2024; 206:114127. [PMID: 38797038 DOI: 10.1016/j.ejca.2024.114127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Homologous Recombination Deficiency (HRD) status predicts response to treatment with poly(ADP-ribose) polymerase inhibitors in Ovarian Cancer (OC) patients. The Myriad myChoiceCDx Assay is approved by Food and Drug Agency for the HRD assessment. Here we compared the HRD status obtained by three commercial panels with the results from Myriad reference test. METHODS The HRD analysis was performed on DNA from formalin-fixed and paraffin-embedded tumor samples of 100 untreated OC patients for which Myriad assay results were available, using TruSight Oncology 500 HRD assay (Illumina), Oncomine Comprehensive Assay Plus (Thermo Fisher Scientific) and SOPHiA DDM HRD solution panel (SOPHiA Genetics). RESULTS A good overall concordance with the reference method was demonstrated at three different levels: BRCA mutational status (from 94.4 % to 97.7 %), the genomic instability value (from 88.2 % to 95.3 %) and for the HRD status (from 90.4 % to 97.6 %). Moreover, a trend in favour of HRD positive patients for response rate, progression-free survival and overall survival similar to Myriad was observed for all three tests. DISCUSSION Our data suggest the feasibility of commercial testing for assessing HRD status, with a good concordance with the reference method and association with clinical outcome.
Collapse
Affiliation(s)
- Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Daniela Califano
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Laura Arenare
- Clinical Trial Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Francesca Bergantino
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Carmela Pisano
- Uro-Gynecological Medical Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Sabrina Chiara Cecere
- Uro-Gynecological Medical Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Giovanni Scambia
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart, Rome, Italy
| | - Domenica Lorusso
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart, Rome, Italy
| | - Grazia Artioli
- Oncologia Medica, ULSS2 Marca Trevigiana, Treviso, Italy
| | - Giulia Tasca
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Anna Spina
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Daniela Russo
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, Naples, Italy
| | | | - Sergio Marchini
- Molecular Pharmacology lab., Group of Cancer Pharmacology IRCCS Humanitas Research Hospital, Rozzano, Milano, Italy
| | - Ettore Domenico Capoluongo
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Naples, Italy; Azienda Ospedaliera per l'Emergenza, Cannizzaro, Catania, Italy
| | - Francesco Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Sandro Pignata
- Uro-Gynecological Medical Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
9
|
Venetis K, Pescia C, Cursano G, Frascarelli C, Mane E, De Camilli E, Munzone E, Dellapasqua S, Criscitiello C, Curigliano G, Guerini Rocco E, Fusco N. The Evolving Role of Genomic Testing in Early Breast Cancer: Implications for Diagnosis, Prognosis, and Therapy. Int J Mol Sci 2024; 25:5717. [PMID: 38891906 PMCID: PMC11172282 DOI: 10.3390/ijms25115717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Multigene prognostic genomic assays have become indispensable in managing early breast cancer (EBC), offering crucial information for risk stratification and guiding adjuvant treatment strategies in conjunction with traditional clinicopathological parameters. The American Society of Clinical Oncology (ASCO) guidelines endorse these assays, though some clinical contexts still lack definitive recommendations. The dynamic landscape of EBC management demands further refinement and optimization of genomic assays to streamline their incorporation into clinical practice. The breast cancer community is poised at the brink of transformative advances in enhancing the clinical utility of genomic assays, aiming to significantly improve the precision and effectiveness of both diagnosis and treatment for women with EBC. This article methodically examines the testing methodologies, clinical validity and utility, costs, diagnostic frameworks, and methodologies of the established genomic tests, including the Oncotype Dx Breast Recurrence Score®, MammaPrint, Prosigna®, EndoPredict®, and Breast Cancer Index (BCI). Among these tests, Prosigna and EndoPredict® have at present been validated only on a prognostic level, while Oncotype Dx, MammaPrint, and BCI hold both a prognostic and predictive role. Oncologists and pathologists engaged in the management of EBC will find in this review a thorough comparison of available genomic assays, as well as strategies to optimize the utilization of the information derived from them.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Eltjona Mane
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisabetta Munzone
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Silvia Dellapasqua
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| |
Collapse
|
10
|
Wehn AK, Qiu P, Lunceford J, Yarunin A, Cristescu R, Liu L, Roessler K, Bilke S, Day JR, Timms KM, Weichert W, Marton MJ. Concordance between an FDA-approved companion diagnostic and an alternative assay kit for assessing homologous recombination deficiency in ovarian cancer. Gynecol Oncol 2024; 184:67-73. [PMID: 38290412 DOI: 10.1016/j.ygyno.2024.01.016] [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: 09/01/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVE Authors evaluated the performance of a commercially available next-generation sequencing assay kit; this was based on genomic content from Illumina's TruSight™ Oncology 500 research assay that identifies BRCA variants and proprietary algorithms licensed from Myriad and, with additional genomic content, measures the homologous recombination deficiency (HRD) genomic instability score (GIS) in tumor tissue (TSO 500 HRD assay). METHODS Data from the TSO 500 HRD assay were compared with data from the Myriad MyChoice®CDx PLUS assay (Myriad assay). Prevalence rates for overall HRD status and BRCA mutations (a deleterious or suspected deleterious BRCA1 or BRCA2 mutation or both) and assay agreement rates for HRD GIS and BRCA analysis were assessed in ovarian tumor samples. Pearson correlations of the continuous HRD GIS and analytic sensitivity and specificity were evaluated. RESULTS The prevalence of overall HRD positivity was 51.2% (TSO 500 HRD assay) versus 49.2% (Myriad assay) and the prevalence of BRCA mutations was 27.6% (TSO 500 HRD assay) versus 25.5% (Myriad assay). After post-processing optimization, concordance of the HRD GIS was 0.980 in all samples and 0.976 in the non-BRCA mutation cohort; the area under the receiver operating characteristic curve was 0.995 and 0.992, respectively. CONCLUSIONS Comparison between the Illumina and Myriad assays showed that overall HRD status, the individual components of BRCA analysis, and HRD GIS detection results were highly concordant (>93%), suggesting the TSO 500 HRD assay will approach the analytical accuracy of the FDA-approved Myriad assay.
Collapse
Affiliation(s)
- Amy K Wehn
- MRL, Merck & Co., Inc., Rahway, NJ, USA.
| | - Ping Qiu
- MRL, Merck & Co., Inc., Rahway, NJ, USA.
| | - Jared Lunceford
- MRL, Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, USA.
| | - Alexander Yarunin
- Scientific Affairs for Global Oncology, AstraZeneca, Milton, Cambridge, United Kingdom.
| | | | - Li Liu
- Assay Development, Illumina, San Diego, CA, USA.
| | | | - Sven Bilke
- Bioinformatics, Illumina, San Diego, CA, USA.
| | - John R Day
- Assay Development, Illumina, San Diego, CA, USA.
| | | | - Wilko Weichert
- Institute of Pathology, Technical University Munich, München, Germany
| | | |
Collapse
|
11
|
Rognoni C, Lorusso D, Costa F, Armeni P. Cost-Effectiveness Analysis of HRD Testing for Previously Treated Patients with Advanced Ovarian Cancer in Italy. Adv Ther 2024; 41:1385-1400. [PMID: 38329713 PMCID: PMC10960911 DOI: 10.1007/s12325-024-02791-3] [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: 11/17/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024]
Abstract
INTRODUCTION Ovarian cancer (OC) is the eighth most common cancer among women, and homologous recombination deficiency (HRD) is present in approximately 50% of these patients. For this group, poly(ADP-ribose) polymerase (PARP) inhibitors are more likely to be effective. The aim of the study was to investigate the cost-effectiveness of HRD testing versus BRCA testing (which identifies mutations present only in 25% of patients) in Italy to optimize the treatment management, possibly with PARP inhibitors. METHODS A cost-effectiveness partition survival model was developed to estimate the expected costs and outcomes (life years, LYs; quality-adjusted life years, QALYs) with lifetime horizon of HRD testing versus BRCA testing alone in women with high-grade serous or endometrioid advanced ovarian cancer. The option to perform the tests in sequence, that is, the BRCA test followed by the HRD test, in patients with BRCA-negative test was also considered, and the model accounted for the National Healthcare Service (NHS) perspective in Italy. The treatments represented the best available options according to the initial test results and according to PARP inhibitors available in Italy. A 3% discount rate was applied. Both deterministic and probabilistic sensitivity analyses were performed to test the robustness of the model results. RESULTS HRD testing was shown to be a cost-effective strategy compared to BRCA testing (incremental cost-utility ratio 22,610€/QALY) and a cost-saving strategy compared to the sequence of tests. The probabilistic sensitivity analysis showed that the HRD test is cost-effective compared to BRCA testing in 98.5% of model simulations considering a willingness-to-pay threshold of 50,000€/QALY. CONCLUSION The identification of genetic anomalies in patients with advanced OC is a costly process. Regardless, HRD upfront testing compared to BRCA testing had a cost-effective profile, allowing the efficient use of healthcare resources and better life expectancy and quality of life for patients.
Collapse
Affiliation(s)
- Carla Rognoni
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Bocconi University, Via Sarfatti 10, 20136, Milan, Italy.
| | - Domenica Lorusso
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | - Francesco Costa
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Bocconi University, Via Sarfatti 10, 20136, Milan, Italy
| | - Patrizio Armeni
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Bocconi University, Via Sarfatti 10, 20136, Milan, Italy
| |
Collapse
|
12
|
Wong RWC, Cheung ANY. Predictive and prognostic biomarkers in female genital tract tumours: an update highlighting their clinical relevance and practical issues. Pathology 2024; 56:214-227. [PMID: 38212229 DOI: 10.1016/j.pathol.2023.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 01/13/2024]
Abstract
The evaluation of biomarkers by molecular techniques and immunohistochemistry has become increasingly relevant to the treatment of female genital tract tumours as a consequence of the greater availability of therapeutic options and updated disease classifications. For ovarian cancer, mutation testing for BRCA1/2 is the standard predictive biomarker for poly(ADP-ribose) polymerase inhibitor therapy, while homologous recombination deficiency testing may allow the identification of eligible patients among cases without demonstrable BRCA1/2 mutations. Clinical recommendations are available which specify how these predictive biomarkers should be applied. Mismatch repair (MMR) protein and folate receptor alpha immunohistochemistry may also be used to guide treatment in ovarian cancer. In endometrial cancer, MMR immunohistochemistry is the preferred test for predicting benefit from immune checkpoint inhibitor (ICI) therapy, but molecular testing for microsatellite instability may have a supplementary role. HER2 testing by immunohistochemistry and in situ hybridisation is applicable to endometrial serous carcinomas to assess trastuzumab eligibility. Immunohistochemistry for oestrogen receptor and progesterone receptor expression may be used for prognostication in endometrial cancer, but its predictive value for hormonal therapy is not yet proven. POLE mutation testing and p53 immunohistochemistry (as a surrogate for TP53 mutation status) serve as prognostic markers for favourable and adverse outcomes, respectively, in endometrial cancer, especially when combined with MMR testing for molecular subtype designation. For cervical cancer, programmed death ligand 1 immunohistochemistry may be used to predict benefit from ICI therapy although its predictive value is under debate. In vulvar cancer, p16 and p53 immunohistochemistry has established prognostic value, stratifying patients into three groups based on the human papillomavirus and TP53 mutation status of the tumour. Awareness of the variety and pitfalls of expression patterns for p16 and p53 in vulvar carcinomas is crucial for accurate designation. It is hoped that collaborative efforts in standardising and optimising biomarker testing for gynaecological tumours will contribute to evidence-based therapeutic decisions.
Collapse
Affiliation(s)
- Richard Wing-Cheuk Wong
- Department of Pathology, United Christian Hospital, Kwun Tong, Hong Kong Special Administrative Region of China.
| | - Annie N Y Cheung
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| |
Collapse
|
13
|
Guarischi-Sousa R, Kroll JE, Bonaldi A, Pierry PM, Villela D, Souza CA, Silva JS, Bürger MC, Oliveira FA, de Paula MG, Meliso FM, de Almeida LG, Monfredini PM, de Oliveira AG, Milanezi F, Scapulatempo-Neto C, Yamamoto GL. A Benchmark of In-House Homologous Recombination Repair Deficiency Testing Solutions for High-Grade Serous Ovarian Cancer Diagnosis. Diagnostics (Basel) 2023; 13:3293. [PMID: 37958189 PMCID: PMC10648202 DOI: 10.3390/diagnostics13213293] [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: 07/17/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 11/15/2023] Open
Abstract
Homologous recombination deficiency (HRD) has become an important prognostic and predictive biomarker for patients with high-grade serous ovarian cancer who may benefit from poly-ADP ribose polymerase inhibitors (PARPi) and platinum-based therapies. HRD testing provides relevant information to personalize patients' treatment options and has been progressively incorporated into diagnostic laboratories. Here, we assessed the performance of an in-house HRD testing system deployable in a diagnostic clinical setting, comparing results from two commercially available next-generation sequencing (NGS)-based tumor tests (SOPHiA DDMTM HRD Solution and AmoyDx® (HRD Focus Panel)) with the reference assay from Myriad MyChoice® (CDx). A total of 85 ovarian cancer samples were subject to HRD testing. An overall strong correlation was observed across the three assays evaluated, regardless of the different underlying methods employed to assess genomic instability, with the highest pairwise correlation between Myriad and SOPHiA (R = 0.87, p-value = 3.39 × 10-19). The comparison of the assigned HRD status to the reference Myriad's test revealed a positive predictive value (PPV) and negative predictive value (NPV) of 90.9% and 96.3% for SOPHiA's test, while AmoyDx's test achieved 75% PPV and 100% NPV. This is the largest HRD testing evaluation using different methodologies and provides a clear picture of the robustness of NGS-based tests currently offered in the market. Our data shows that the implementation of in-house HRD testing in diagnostic laboratories is technically feasible and can be reliably performed with commercial assays. Also, the turnaround time is compatible with clinical needs, making it an ideal alternative to offer to a broader number of patients while maintaining high-quality standards at more accessible price tiers.
Collapse
|