227 results on '"Llop-Guevara A"'
Search Results
202. A RAD51 functional assay as a candidate test for homologous recombination deficiency in ovarian cancer.
- Author
-
Blanc-Durand, Félix, Yaniz-Galende, Elisa, Llop-Guevara, Alba, Genestie, Catherine, Serra, Violeta, Herencia-Ropero, Andrea, Klein, Christophe, Berton, Dominique, Lortholary, Alain, Dohollou, Nadine, Desauw, Christophe, Fabbro, Michel, Malaurie, Emmanuelle, Bonichon-Lamaichhane, Nathalie, Dubot, Coraline, Kurtz, Jean Emmanuel, de Rauglaudre, Gaëtan, Raban, Nadia, Chevalier-Place, Annick, and Ferron, Gwenael
- Subjects
- *
OVARIAN cancer , *OSTEOCHONDROSIS , *NEOADJUVANT chemotherapy , *DNA damage , *PROGRESSION-free survival , *PLATINUM - Abstract
Homologous recombination deficiency (HRD), defined as BRCA1/2 mutation (BRCAmut) or high genomic instability, is used to identify ovarian cancer (OC) patients most likely to benefit from PARP inhibitors. While these tests are useful, they are imperfect. Another approach is to measure the capacity of tumor cells to form RAD51 foci in the presence of DNA damage using an immunofluorescence assay (IF). We aimed to describe for the first time this assay in OC and correlate it to platinum response and BRCAmut. Tumor samples were prospectively collected from the randomized CHIVA trial of neoadjuvant platinum +/− nintedanib. IF for RAD51, GMN and gH2AX was performed on FFPE blocks. Tumors were considered RAD51-low if ≤10% of GMN-positive tumor cells had ≥5 RAD51 foci. BRCAmut were identified by NGS. 155 samples were available. RAD51 assay was contributive for 92% of samples and NGS available for 77%. gH2AX foci confirmed the presence of significant basal DNA damage. 54% of samples were considered HRD by RAD51 and presented higher overall response rates to neoadjuvant platinum (P = 0.04) and longer progression-free survival (P = 0.02). In addition, 67% of BRCAmut were HRD by RAD51. Among BRCAmut, RAD51-high tumors seem to harbor poorer response to chemotherapy (P = 0.02). We evaluated a functional assay of HR competency. OC demonstrate high levels of DNA damage, yet 54% fail to form RAD51 foci. These RAD51-low OC tend to be more sensitive to neoadjuvant platinum. The RAD51 assay also identified a subset of RAD51-high BRCAmut tumors with unexpected poor platinum response. • RAD51 functional assay requires minimal tissue and yields contributive results in >90% of cases. • This assay identifies 54% of EOC patients with RAD51-low tumors and these patients presented better outcome. • Among BRCA-mutated tumors, RAD51-high tumors showed poorer response to neoadjuvant chemotherapy [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
203. A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation
- Author
-
Castroviejo-Bermejo, Marta, Cruz, Cristina, Llop-Guevara, Alba, Gutiérrez-Enríquez, Sara, Ducy, Mandy, Ibrahim, Yasir Hussein, Gris-Oliver, Albert, Pellegrino, Benedetta, Bruna, Alejandra, Guzmán, Marta, Rodríguez, Olga, Grueso, Judit, Bonache, Sandra, Moles-Fernández, Alejandro, Villacampa, Guillermo, Viaplana, Cristina, Gómez, Patricia, Vidal, Maria, Peg, Vicente, Serres-Créixams, Xavier, Dellaire, Graham, Simard, Jacques, Nuciforo, Paolo, Rubio, Isabel T, Dienstmann, Rodrigo, Barrett, J Carl, Caldas, Carlos, Baselga, José, Saura, Cristina, Cortés, Javier, Déas, Olivier, Jonkers, Jos, Masson, Jean-Yves, Cairo, Stefano, Judde, Jean-Gabriel, O'Connor, Mark J, Díez, Orland, Balmaña, Judith, and Serra, Violeta
- Subjects
homologous recombination ,Antineoplastic Agents ,Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,BRCA1 ,Piperazines ,3. Good health ,Mice ,Drug Resistance, Neoplasm ,PALB2 ,Biomarkers, Tumor ,RAD51 ,Animals ,Heterografts ,Humans ,Phthalazines ,Female ,Rad51 Recombinase ,PARP inhibitors - Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are effective in cancers with defective homologous recombination DNA repair (HRR), including BRCA1/2-related cancers. A test to identify additional HRR-deficient tumors will help to extend their use in new indications. We evaluated the activity of the PARPi olaparib in patient-derived tumor xenografts (PDXs) from breast cancer (BC) patients and investigated mechanisms of sensitivity through exome sequencing, BRCA1 promoter methylation analysis, and immunostaining of HRR proteins, including RAD51 nuclear foci. In an independent BC PDX panel, the predictive capacity of the RAD51 score and the homologous recombination deficiency (HRD) score were compared. To examine the clinical feasibility of the RAD51 assay, we scored archival breast tumor samples, including PALB2-related hereditary cancers. The RAD51 score was highly discriminative of PARPi sensitivity versus PARPi resistance in BC PDXs and outperformed the genomic test. In clinical samples, all PALB2-related tumors were classified as HRR-deficient by the RAD51 score. The functional biomarker RAD51 enables the identification of PARPi-sensitive BC and broadens the population who may benefit from this therapy beyond BRCA1/2-related cancers.
204. Additional file 3: of Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Llop-Guevara, Alba, Porras, Mónica, Cendón, Carla, Ceglie, Irene Di, Siracusa, Francesco, Madarena, Federica, Vagelis Rinotas, Lluís Gómez, Lent, Peter Van, Douni, Eleni, Chang, Hyun Dong, Kamradt, Thomas, and Román, Juan
- Subjects
3. Good health - Abstract
Effect of Janus kinase inhibitor ( JAKi )/spleen tyrosine kinase inhibitor ( SYKi ) treatment on body weight and cell viability. a DBA/1 mice were immunized with glucose-6-phosphate isomerase (G6PI) + complete Freund’s adjuvant (CFA) on day 0. b CD-1 mice were immunized with keyhole limpet hemocyanin (KLH) on day 14 (T cell-dependent antibody response (TDAR) assay). a, b Relative body weight to day 0 (before treatment started) was recorded regularly. As expected, naive animals progressively gained weight over time and treatment with the glucocorticoid prednisone induced the most significant decrease in body weight. G6PI immunization triggered the development of arthritis and, therefore, vehicle-treated animals initially lost weight and then slowly recovered. Modest changes were observed in vehicle-exposed mice upon KLH immunization. Body weight in mice daily treated with single or dual JAK/SYK inhibition was comparable to, or even better than, vehicle-treated animals, clearly showing that treatment with JAKi + SYKi did not negatively impact the general wellbeing of mice. c HepG2 cells were exposed for 72 h to increasing doses of JAKi/SYKi (0.5, 5 or 50 μM) or diclofenac (positive control of hepatotoxicity) and mitochondrial activity was evaluated by Alamar Blue fluorescence. JAKi did not show any cytotoxic effect at the assayed concentrations. SYKi significantly decreased cell viability only at the highest dose (50 μM). Importantly, plasma levels of SYKi peaked at 3–4 μM and for a very short time (see Additional file 1e); therefore, higher and sustained concentrations are never reached in vivo. These data show that combined inhibition of JAK + SYK does not result in higher general toxicity. Graphs show mean (± standard error of the mean) for 3–6 mice/group or 2–10 replicates/condition, *p
205. Additional file 3: of Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Llop-Guevara, Alba, Porras, Mónica, Cendón, Carla, Ceglie, Irene Di, Siracusa, Francesco, Madarena, Federica, Vagelis Rinotas, Lluís Gómez, Lent, Peter Van, Douni, Eleni, Chang, Hyun Dong, Kamradt, Thomas, and Román, Juan
- Subjects
3. Good health - Abstract
Effect of Janus kinase inhibitor ( JAKi )/spleen tyrosine kinase inhibitor ( SYKi ) treatment on body weight and cell viability. a DBA/1 mice were immunized with glucose-6-phosphate isomerase (G6PI) + complete Freund’s adjuvant (CFA) on day 0. b CD-1 mice were immunized with keyhole limpet hemocyanin (KLH) on day 14 (T cell-dependent antibody response (TDAR) assay). a, b Relative body weight to day 0 (before treatment started) was recorded regularly. As expected, naive animals progressively gained weight over time and treatment with the glucocorticoid prednisone induced the most significant decrease in body weight. G6PI immunization triggered the development of arthritis and, therefore, vehicle-treated animals initially lost weight and then slowly recovered. Modest changes were observed in vehicle-exposed mice upon KLH immunization. Body weight in mice daily treated with single or dual JAK/SYK inhibition was comparable to, or even better than, vehicle-treated animals, clearly showing that treatment with JAKi + SYKi did not negatively impact the general wellbeing of mice. c HepG2 cells were exposed for 72 h to increasing doses of JAKi/SYKi (0.5, 5 or 50 μM) or diclofenac (positive control of hepatotoxicity) and mitochondrial activity was evaluated by Alamar Blue fluorescence. JAKi did not show any cytotoxic effect at the assayed concentrations. SYKi significantly decreased cell viability only at the highest dose (50 μM). Importantly, plasma levels of SYKi peaked at 3–4 μM and for a very short time (see Additional file 1e); therefore, higher and sustained concentrations are never reached in vivo. These data show that combined inhibition of JAK + SYK does not result in higher general toxicity. Graphs show mean (± standard error of the mean) for 3–6 mice/group or 2–10 replicates/condition, *p
206. Additional file 1: of Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Llop-Guevara, Alba, Porras, Mónica, Cendón, Carla, Ceglie, Irene Di, Siracusa, Francesco, Madarena, Federica, Vagelis Rinotas, Lluís Gómez, Lent, Peter Van, Douni, Eleni, Chang, Hyun Dong, Kamradt, Thomas, and Román, Juan
- Subjects
3. Good health - Abstract
Potency, selectivity and pharmacokinetics of tofacitinib/CP-690,550 (Janus kinase inhibitor ( JAKi )) and PRT-062607 (spleen tyrosine kinase inhibitor ( SYKi )). a, b Human cell lines were pre-incubated with increasing concentrations of JAKi or SYKi (0.005 to 5 μM) for 1 h and then stimulated with either 100 ng/ml rmIL-6 for 15 minutes or 10 μg/ml anti-IgM for 5 minutes. Cells were fixed, permeabilized, stained with anti-STAT3(pY705)-Alexa Fluor 647 or anti-BLNK(pY84)-Alexa Fluor 488 and quantified by flow cytometry. Half maximal inhibitory concentration (IC 50 ) was extrapolated from an inhibition vs log (concentration) curve. c Collagen-induced arthritis (CIA) model. DBA/1 mice were immunized on day 0 with type II collagen (100 μg/mouse, subcutaneous administration (s.c.)) emulsified in complete Freund’s adjuvant (CFA) and boosted on day 21. Starting on day 30, when disease was mild, mice were orally treated once a day with increasing doses of JAKi (3.75 to 30 mg/kg). Graph shows mean for 6–8 mice/group. d Collagen antibody-induced arthritis (CAIA) model. BALB/c mice received a cocktail of monoclonal antibodies (2 mg, intravenous administration (i.v.)) directed against type II collagen on day 0 and a lipolysaccharide challenge (70 μg, intraperitoneal administration (i.p.)) on day 3. Twice daily treatment with 30 mg/kg SYKi started on day 4. Graph shows mean for 6–8 mice/group. For monitoring both CIA and CAIA models, each paw was scored individually on a scale of 0–4, with 4 indicating the most severe swelling and erythema. e DBA/1 mice received a single oral dose of JAKi (20 mg/kg) or SYKi (30 mg/kg) and plasmatic drug concentration was determined by liquid chromatography-tandem mass spectrometry. Graph shows mean for 2–7 mice/group and a line representing IC50 values obtained in a and b. (PDF 755 kb)
207. Additional file 2: of Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Llop-Guevara, Alba, Porras, Mónica, Cendón, Carla, Ceglie, Irene Di, Siracusa, Francesco, Madarena, Federica, Vagelis Rinotas, Lluís Gómez, Lent, Peter Van, Douni, Eleni, Chang, Hyun Dong, Kamradt, Thomas, and Román, Juan
- Subjects
3. Good health - Abstract
Clinical and immunological comparison of acute versus chronic glucose-6-phosphate isomerase ( G6PI )-induced arthritis model in mice. a Diagram summarizing the experimental protocol followed in Figs. 1, 2, 3, 4 and 5 (see “Methods” for more details). b Time course for arthritis scores clearly showing moderate and acute arthritis in G6PI-immunized mice, and severe and chronic arthritis in T regulatory cell (Treg)-depleted and G6PI-immunized mice. c Proportion of CD25hiFoxp3+ Treg cells in CD3+CD4+ cells at the time of immunization (day 0). d Numbers of total cells from inguinal lymph nodes and e leucocyte counts in peripheral blood at day 6 post-immunization. f Cytokine secretion by splenocytes isolated on day 31 and stimulated in vitro with 20 μg/ml G6PI for 72 h. Graphs show mean (± standard error of the mean) for 3–6 mice/group, *p
208. Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Francesco Siracusa, Lluis A. Gomez, Hyun-Dong Chang, Alba Llop-Guevara, Monica Porras, Carla Cendón, Peter L E M van Lent, Juan Román, Eleni Douni, Thomas Kamradt, Vagelis Rinotas, Federica Madarena, and Irene Di Ceglie
- Subjects
Male ,Small molecule inhibitors ,medicine.medical_treatment ,Syk ,Arthritis ,Osteoclasts ,Pharmacology ,Etanercept ,Arthritis, Rheumatoid ,Mice ,0302 clinical medicine ,Piperidines ,Medicine ,SYK ,Immunology and Allergy ,Memory cells ,Fibroblast-like synoviocytes ,0303 health sciences ,Cyclohexylamines ,Mice, Inbred BALB C ,Kinase ,Anti-TNF therapy ,Intracellular Signaling Peptides and Proteins ,Protein-Tyrosine Kinases ,Flow Cytometry ,3. Good health ,Cytokine ,Mice, Inbred DBA ,Antirheumatic Agents ,Drug Therapy, Combination ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] ,medicine.drug ,Research Article ,Immunology ,Enzyme-Linked Immunosorbent Assay ,Immunophenotyping ,03 medical and health sciences ,Rheumatology ,Animals ,Syk Kinase ,Pyrroles ,Protein Kinase Inhibitors ,030304 developmental biology ,Janus Kinases ,030203 arthritis & rheumatology ,Tofacitinib ,business.industry ,medicine.disease ,Arthritis, Experimental ,JAK ,Destructive Arthritis ,Disease Models, Animal ,Pyrimidines ,business ,Janus kinase - Abstract
Introduction Despite the broad spectrum of antirheumatic drugs, RA is still not well controlled in up to 30-50 % of patients. Inhibition of JAK kinases by means of the pan-JAK inhibitor tofacitinib has demonstrated to be effective even in difficult-to-treat patients. Here, we discuss whether the efficacy of JAK inhibition can be improved by simultaneously inhibiting SYK kinase, since both kinases mediate complementary and non-redundant pathways in RA. Methods Efficacy of dual JAK + SYK inhibition with selective small molecule inhibitors was evaluated in chronic G6PI-induced arthritis, a non-self-remitting and destructive arthritis model in mice. Clinical and histopathological scores, as well as cytokine and anti-G6PI antibody production were assessed in both preventive and curative protocols. Potential immunotoxicity was also evaluated in G6PI-induced arthritis and in a 28-day TDAR model, by analysing the effects of JAK + SYK inhibition on hematological parameters, lymphoid organs, leukocyte subsets and cell function. Results Simultaneous JAK + SYK inhibition completely prevented mice from developing arthritis. This therapeutic strategy was also very effective in ameliorating already established arthritis. Dual kinase inhibition immediately resulted in greatly decreased clinical and histopathological scores and led to disease remission in over 70 % of the animals. In contrast, single JAK inhibition and anti-TNF therapy (etanercept) were able to stop disease progression but not to revert it. Dual kinase inhibition decreased Treg and NK cell counts to the same extent as single JAK inhibition but overall cytotoxicity remained intact. Interestingly, treatment discontinuation rapidly reversed such immune cell reduction without compromising clinical efficacy, suggesting long-lasting curative effects. Dual kinase inhibition reduced the Th1/Th17 cytokine cascade and the differentiation and function of joint cells, in particular osteoclasts and fibroblast-like synoviocytes. Conclusions Concurrent JAK + SYK inhibition resulted in higher efficacy than single kinase inhibition and TNF blockade in a chronic and severe arthritis model. Thus, blockade of multiple immune signals with dual JAK + SYK inhibition represents a reasonable therapeutic strategy for RA, in particular in patients with inadequate responses to current treatments. Our data supports the multiplicity of events underlying this heterogeneous and complex disease. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0866-0) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
209. Additional file 2: of Simultaneous inhibition of JAK and SYK kinases ameliorates chronic and destructive arthritis in mice
- Author
-
Llop-Guevara, Alba, Porras, Mónica, Cendón, Carla, Ceglie, Irene Di, Siracusa, Francesco, Madarena, Federica, Vagelis Rinotas, Lluís Gómez, Lent, Peter Van, Douni, Eleni, Chang, Hyun Dong, Kamradt, Thomas, and Román, Juan
- Subjects
3. Good health - Abstract
Clinical and immunological comparison of acute versus chronic glucose-6-phosphate isomerase ( G6PI )-induced arthritis model in mice. a Diagram summarizing the experimental protocol followed in Figs. 1, 2, 3, 4 and 5 (see “Methods” for more details). b Time course for arthritis scores clearly showing moderate and acute arthritis in G6PI-immunized mice, and severe and chronic arthritis in T regulatory cell (Treg)-depleted and G6PI-immunized mice. c Proportion of CD25hiFoxp3+ Treg cells in CD3+CD4+ cells at the time of immunization (day 0). d Numbers of total cells from inguinal lymph nodes and e leucocyte counts in peripheral blood at day 6 post-immunization. f Cytokine secretion by splenocytes isolated on day 31 and stimulated in vitro with 20 μg/ml G6PI for 72 h. Graphs show mean (± standard error of the mean) for 3–6 mice/group, *p
210. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer
- Author
-
Cruz, C, Castroviejo-Bermejo, M, Gutiérrez-Enríquez, S, Llop-Guevara, A, Ibrahim, YH, Gris-Oliver, A, Bonache, S, Morancho, B, Bruna, A, Rueda, OM, Lai, Z, Polanska, UM, Jones, GN, Kristel, P, De Bustos, L, Guzman, M, Rodríguez, O, Grueso, J, Montalban, G, Caratú, G, Mancuso, F, Fasani, R, Jiménez, J, Howat, WJ, Dougherty, B, Vivancos, A, Nuciforo, P, Serres-Créixams, X, Rubio, IT, Oaknin, A, Cadogan, E, Barrett, JC, Caldas, C, Baselga, J, Saura, C, Cortés, J, Arribas, J, Jonkers, J, Díez, O, O'Connor, MJ, Balmaña, J, and Serra, V
- Subjects
BRCA2 Protein ,BRCA1 Protein ,Mice, Nude ,Recombinational DNA Repair ,Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,Xenograft Model Antitumor Assays ,3. Good health ,Mice ,Treatment Outcome ,Drug Resistance, Neoplasm ,Biomarkers, Tumor ,Animals ,Humans ,Female ,Breast ,Rad51 Recombinase ,Germ-Line Mutation ,Retrospective Studies - Abstract
BACKGROUND: BRCA1 and BRCA2 (BRCA1/2)-deficient tumors display impaired homologous recombination repair (HRR) and enhanced sensitivity to DNA damaging agents or to poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi). Their efficacy in germline BRCA1/2 (gBRCA1/2)-mutated metastatic breast cancers has been recently confirmed in clinical trials. Numerous mechanisms of PARPi resistance have been described, whose clinical relevance in gBRCA-mutated breast cancer is unknown. This highlights the need to identify functional biomarkers to better predict PARPi sensitivity. PATIENTS AND METHODS: We investigated the in vivo mechanisms of PARPi resistance in gBRCA1 patient-derived tumor xenografts (PDXs) exhibiting differential response to PARPi. Analysis included exome sequencing and immunostaining of DNA damage response proteins to functionally evaluate HRR. Findings were validated in a retrospective sample set from gBRCA1/2-cancer patients treated with PARPi. RESULTS: RAD51 nuclear foci, a surrogate marker of HRR functionality, were the only common feature in PDX and patient samples with primary or acquired PARPi resistance. Consistently, low RAD51 was associated with objective response to PARPi. Evaluation of the RAD51 biomarker in untreated tumors was feasible due to endogenous DNA damage. In PARPi-resistant gBRCA1 PDXs, genetic analysis found no in-frame secondary mutations, but BRCA1 hypomorphic proteins in 60% of the models, TP53BP1-loss in 20% and RAD51-amplification in one sample, none mutually exclusive. Conversely, one of three PARPi-resistant gBRCA2 tumors displayed BRCA2 restoration by exome sequencing. In PDXs, PARPi resistance could be reverted upon combination of a PARPi with an ataxia-telangiectasia mutated (ATM) inhibitor. CONCLUSION: Detection of RAD51 foci in gBRCA tumors correlates with PARPi resistance regardless of the underlying mechanism restoring HRR function. This is a promising biomarker to be used in the clinic to better select patients for PARPi therapy. Our study also supports the clinical development of PARPi combinations such as those with ATM inhibitors.
211. A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation.
- Author
-
Castroviejo‐Bermejo, Marta, Cruz, Cristina, Llop‐Guevara, Alba, Gutiérrez‐Enríquez, Sara, Ducy, Mandy, Ibrahim, Yasir Hussein, Gris‐Oliver, Albert, Pellegrino, Benedetta, Bruna, Alejandra, Guzmán, Marta, Rodríguez, Olga, Grueso, Judit, Bonache, Sandra, Moles‐Fernández, Alejandro, Villacampa, Guillermo, Viaplana, Cristina, Gómez, Patricia, Vidal, Marc, Peg, Vicente, and Serres‐Créixams, Xavier
- Abstract
Poly(ADP‐ribose) polymerase (PARP) inhibitors (PARPi) are effective in cancers with defective homologous recombination DNA repair (HRR), including BRCA1/2‐related cancers. A test to identify additional HRR‐deficient tumors will help to extend their use in new indications. We evaluated the activity of the PARPi olaparib in patient‐derived tumor xenografts (PDXs) from breast cancer (BC) patients and investigated mechanisms of sensitivity through exome sequencing, BRCA1 promoter methylation analysis, and immunostaining of HRR proteins, including RAD51 nuclear foci. In an independent BC PDX panel, the predictive capacity of the RAD51 score and the homologous recombination deficiency (HRD) score were compared. To examine the clinical feasibility of the RAD51 assay, we scored archival breast tumor samples, including PALB2‐related hereditary cancers. The RAD51 score was highly discriminative of PARPi sensitivity versus PARPi resistance in BC PDXs and outperformed the genomic test. In clinical samples, all PALB2‐related tumors were classified as HRR‐deficient by the RAD51 score. The functional biomarker RAD51 enables the identification of PARPi‐sensitive BC and broadens the population who may benefit from this therapy beyond BRCA1/2‐related cancers. Synopsis: Sensitive and highly specific biomarkers usable in archived formalin fixed parafin embedded (FFPE) tumour samples are needed to extend the use of PARP inhibitors beyond BRCA1/2‐related cancers. The RAD51 score may satisfy this clinical unmet need. The RAD51 score shows complete discriminative capacity in predicting PARP inhibitor response.The RAD51 score is feasible in routine breast tumor samples without prior exposure to DNA damaging agents.Carrying a PALB2 mutation is associated with a low RAD51score. Sensitive and highly specific biomarkers usable in archived formalin fixed parafin embedded (FFPE) tumour samples are needed to extend the use of PARP inhibitors beyond BRCA1/2‐related cancers. The RAD51 score may satisfy this clinical unmet need. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
212. Homologous Recombination Deficiency Across Subtypes of Primary Breast Cancer.
- Author
-
Yndestad, Synnøve, Engebrethsen, Christina, Herencia-Ropero, Andrea, Nikolaienko, Oleksii, Vintermyr, Olav K., Lillestøl, Reidun K., Minsaas, Laura, Leirvaag, Beryl, Iversen, Gjertrud T., Gilje, Bjørnar, Blix, Egil S., Espelid, Helge, Lundgren, Steinar, Geisler, Jürgen, Aase, Hildegunn S., Aas, Turid, Gudlaugsson, Einar G., Llop-Guevara, Alba, Serra, Violeta, and Janssen, Emiel A.M.
- Subjects
- *
HOMOLOGOUS recombination , *BREAST cancer , *CANCER patients , *TRIPLE-negative breast cancer , *HORMONE receptor positive breast cancer , *TUMOR-infiltrating immune cells - Abstract
PURPOSE: Homologous recombination deficiency (HRD) is highly prevalent in triple-negative breast cancer (TNBC) and associated with response to PARP inhibition (PARPi). Here, we studied the prevalence of HRD in non-TNBC to assess the potential for PARPi in a wider group of patients with breast cancer. METHODS: HRD status was established using targeted gene panel sequencing (360 genes) and BRCA1 methylation analysis of pretreatment biopsies from 201 patients with primary breast cancer in the phase II PETREMAC trial (ClinicalTrials.gov identifier: NCT02624973). HRD was defined as mutations in BRCA1 , BRCA2 , BRIP1 , BARD1 , or PALB2 and/or promoter methylation of BRCA1 (strict definition; HRD-S). In secondary analyses, a wider definition (HRD-W) was used, examining mutations in 20 additional genes. Furthermore, tumor BRCA ness (multiplex ligation-dependent probe amplification), PAM50 subtyping, RAD51 nuclear foci to test functional HRD, tumor-infiltrating lymphocyte (TIL), and PD-L1 analyses were performed. RESULTS: HRD-S was present in 5% of non-TNBC cases (n = 9 of 169), contrasting 47% of the TNBC tumors (n = 15 of 32). HRD-W was observed in 23% of non-TNBC (n = 39 of 169) and 59% of TNBC cases (n = 19 of 32). Of 58 non-TNBC and 30 TNBC biopsies examined for RAD51 foci, 4 of 4 (100%) non-TNBC and 13 of 14 (93%) TNBC cases classified as HRD-S had RAD51 low scores. In contrast, 4 of 17 (24%) non-TNBC and 15 of 19 (79%) TNBC biopsies classified as HRD-W exhibited RAD51 low scores. Of nine non-TNBC tumors with HRD-S status, only one had a basal-like PAM50 signature. There was a high concordance between HRD-S and either BRCA ness, high TIL density, or high PD-L1 expression (each P <.001). CONCLUSION: The prevalence of HRD in non-TNBC suggests that therapy targeting HRD should be evaluated in a wider breast cancer patient population. Strict HRD criteria should be implemented to increase diagnostic precision with respect to functional HRD. Some non–triple-negative breast cancers are HRD and could be considered for PARP inhibition therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
213. Corrigendum to "A RAD51 functional assay as a candidate test for homologous recombination deficiency in ovarian cancer".
- Author
-
Blanc-Durand, Félix, Yaniz-Galende, Elisa, Llop-Guevara, Alba, Genestie, Catherine, Serra, Violeta, Herencia-Ropero, Andrea, Klein, Christophe, Berton, Dominique, Lortholary, Alain, Dohollou, Nadine, Desauw, Christophe, Fabbro, Michel, Malaurie, Emmanuelle, Bonichon-Lamaichhane, Nathalie, Dubot, Coraline, Kurtz, Jean Emmanuel, de Rauglaudre, Gaëtan, Raban, Nadia, Chevalier-Place, Annick, and Ferron, Gwenael
- Subjects
- *
OVARIAN cancer - Published
- 2023
- Full Text
- View/download PDF
214. Basal expression of RAD51 foci predicts olaparib response in patient-derived ovarian cancer xenografts.
- Author
-
Guffanti, F., Alvisi, M F, Anastasia, A., Ricci, F., Chiappa, M., Llop-Guevara, A., Serra, V., Fruscio, R., Degasperi, A., Nik-Zainal, S., Bani, M R, Lupia, M., Giavazzi, R., Rulli, E., and Damia, G.
- Abstract
Background: The search for biomarkers to evaluate ovarian cancer (OC) homologous recombination (HR) function and predict the response to therapy is an urgent clinical need to improve the selection of patients who could benefit from platinum- and olaparib (poly-ADP ribose polymerase inhibitors, PARPi)-based therapies.Methods: We used a large collection of OC patient-derived xenografts (PDXs) (n = 47) and evaluated their HR status based on BRCA1/2 mutations, BRCA1 promoter methylation and the HRDetect score. RAD51 foci were quantified in formalin-fixed, paraffin-embedded untreated tumour specimens by immunofluorescence and the messenger RNA expression of 21 DNA repair genes by real-time PCR.Results: Tumour HR deficiency predicted both platinum and olaparib responses. The basal level of RAD51 foci evaluated in geminin-positive/replicating cells strongly inversely correlated with olaparib response (p = 0.011); in particular, the lower the foci score, the greater the sensitivity to olaparib, while low RAD51 foci score seems to associate with platinum activity.Conclusions: The basal RAD51 foci score is a candidate predictive biomarker of olaparib response in OC patients as it can be easily translatable in a clinical setting. Moreover, the findings corroborate the importance of OC-PDXs as a reliable tool to identify and validate biomarkers of response to therapy. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
215. Olaparib monotherapy as primary treatment in unselected triple negative breast cancer.
- Author
-
Eikesdal, H.P., Yndestad, S., Elzawahry, A., Llop-Guevara, A., Gilje, B., Blix, E.S., Espelid, H., Lundgren, S., Geisler, J., Vagstad, G., Venizelos, A., Minsaas, L., Leirvaag, B., Gudlaugsson, E.G., Vintermyr, O.K., Aase, H.S., Aas, T., Balmaña, J., Serra, V., and Janssen, E.A.M.
- Subjects
- *
BREAST cancer treatment , *ANTINEOPLASTIC agents , *ADENOSINE diphosphate ribose , *BRCA genes , *TRIPLE-negative breast cancer , *CANCER genetics , *NUCLEOTIDE sequence - Abstract
The antitumor efficacy of PARP inhibitors (PARPi) for breast cancer patients harboring germline BRCA1/2 (g BRCA1/2) mutations is well established. While PARPi monotherapy was ineffective in patients with metastatic triple negative breast cancer (TNBC) wild type for BRCA1/2 , we hypothesized that PARPi may be effective in primary TNBCs without previous chemotherapy exposure. In the phase II PETREMAC trial, patients with primary TNBC >2 cm received olaparib for up to 10 weeks before chemotherapy. Tumor biopsies collected before and after olaparib underwent targeted DNA sequencing (360 genes) and BRCA1 methylation analyses. In addition, BRCA ness (multiplex ligation-dependent probe amplification), PAM50 gene expression, RAD51 foci, tumor-infiltrating lymphocytes (TILs) and PD-L1 analyses were performed on pretreatment samples. The median pretreatment tumor diameter was 60 mm (range 25-112 mm). Eighteen out of 32 patients obtained an objective response (OR) to olaparib (56.3%). Somatic or germline mutations affecting homologous recombination (HR) were observed in 10/18 responders [OR 55.6%, 95% confidence interval (CI) 33.7-75.4] contrasting 1/14 non-responders (OR 7.1%; CI 1.3-31.5, P = 0.008). Among tumors without HR mutations, 6/8 responders versus 3/13 non-responders revealed BRCA1 hypermethylation (P = 0.03). Thus, 16/18 responders (88.9%, CI 67.2-96.9), in contrast to 4/14 non-responders (28.6%, CI 11.7-54.7, P = 0.0008), carried HR mutations and/or BRCA1 methylation. Excluding one g PALB2 and four g BRCA1/ 2 mutation carriers, 12/14 responders (85.7%, CI 60.1-96.0) versus 3/13 non-responders (23.1%, CI 8.2-50.3, P = 0.002) carried somatic HR mutations and/or BRCA1 methylation. In contrast to BRCA ness signature or basal-like subtype, low RAD51 scores, high TIL or high PD-L1 expression all correlated to olaparib response. Olaparib yielded a high clinical response rate in treatment-naïve TNBCs revealing HR deficiency, beyond germline HR mutations. ClinicalTrials.gov identifier: NCT02624973. • We observed an objective response to olaparib in 18 out of 32 (56%) of unselected, primary triple negative breast cancers. • Homologous recombination deficiency (HRD) was determined by targeted DNA sequencing and BRCA1 methylation. • HRD was predictive of response to olaparib and present in 16 out of 18 responders. • HRD was also predictive of response to olaparib beyond germline BRCA1/2 and g PALB2 mutations; 12 out of 14 responders. • Olaparib was associated with minor side-effects and did not influence subsequent tolerance to chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
216. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer.
- Author
-
Cruz, C, Castroviejo-Bermejo, M, Gutiérrez-Enríquez, S, Llop-Guevara, A, Ibrahim, Y H, Gris-Oliver, A, Bonache, S, Morancho, B, Bruna, A, and Rueda, O M
- Subjects
- *
GERM cells , *BREAST cancer treatment , *HOMOLOGOUS chromosomes , *CANCER chemotherapy , *XENOGRAFTS - Abstract
Background: BRCA1 and BRCA2 (BRCA1/2)-deficient tumors display impaired homologous recombination repair (HRR) and enhanced sensitivity to DNA damaging agents or to poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi). Their efficacy in germline BRCA1/2 (gBRCA1/2)-mutated metastatic breast cancers has been recently confirmed in clinical trials. Numerous mechanisms of PARPi resistance have been described, whose clinical relevance in gBRCA-mutated breast cancer is unknown. This highlights the need to identify functional biomarkers to better predict PARPi sensitivity. Patients and methods: We investigated the in vivo mechanisms of PARPi resistance in gBRCA1 patient-derived tumor xenografts (PDXs) exhibiting differential response to PARPi. Analysis included exome sequencing and immunostaining of DNA damage response proteins to functionally evaluate HRR. Findings were validated in a retrospective sample set from gBRCA1/ 2-cancer patients treated with PARPi. Results: RAD51 nuclear foci, a surrogate marker of HRR functionality, were the only common feature in PDX and patient samples with primary or acquired PARPi resistance. Consistently, low RAD51 was associated with objective response to PARPi. Evaluation of the RAD51 biomarker in untreated tumors was feasible due to endogenous DNA damage. In PARPi-resistant gBRCA1 PDXs, genetic analysis found no in-frame secondary mutations, but BRCA1 hypomorphic proteins in 60% of the models, TP53BP1-loss in 20% and RAD51-amplification in one sample, none mutually exclusive. Conversely, one of three PARPi-resistant gBRCA2 tumors displayed BRCA2 restoration by exome sequencing. In PDXs, PARPi resistance could be reverted upon combination of a PARPi with an ataxia-telangiectasia mutated (ATM) inhibitor. Conclusion: Detection of RAD51 foci in gBRCA tumors correlates with PARPi resistance regardless of the underlying mechanism restoring HRR function. This is a promising biomarker to be used in the clinic to better select patients for PARPi therapy. Our study also supports the clinical development of PARPi combinations such as those with ATM inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
217. A living biobank of patient-derived ductal carcinoma in situ mouse-intraductal xenografts identifies risk factors for invasive progression.
- Author
-
Hutten, Stefan J., de Bruijn, Roebi, Lutz, Catrin, Badoux, Madelon, Eijkman, Timo, Chao, Xue, Ciwinska, Marta, Sheinman, Michael, Messal, Hendrik, Herencia-Ropero, Andrea, Kristel, Petra, Mulder, Lennart, van der Waal, Rens, Sanders, Joyce, Almekinders, Mathilde M., Llop-Guevara, Alba, Davies, Helen R., van Haren, Matthijs J., Martin, Nathaniel I., and Behbod, Fariba
- Subjects
- *
CARCINOMA in situ , *DUCTAL carcinoma , *XENOGRAFTS , *PROGNOSIS , *CANCER invasiveness - Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer (IBC). Due to a lack of biomarkers able to distinguish high- from low-risk cases, DCIS is treated similar to early IBC even though the minority of untreated cases eventually become invasive. Here, we characterized 115 patient-derived mouse-intraductal (MIND) DCIS models reflecting the full spectrum of DCIS observed in patients. Utilizing the possibility to follow the natural progression of DCIS combined with omics and imaging data, we reveal multiple prognostic factors for high-risk DCIS including high grade, HER2 amplification, expansive 3D growth, and high burden of copy number aberrations. In addition, sequential transplantation of xenografts showed minimal phenotypic and genotypic changes over time, indicating that invasive behavior is an intrinsic phenotype of DCIS and supporting a multiclonal evolution model. Moreover, this study provides a collection of 19 distributable DCIS-MIND models spanning all molecular subtypes. [Display omitted] • Development of a biobank of DCIS-MIND models retaining primary features • Identification of multiple factors associated with invasive progression of DCIS • Identification of two distinct 3D growth patterns associated with outcome • Development of a collection of 19 distributable DCIS models Hutten et al. generate 115 DCIS-MIND models and characterize them with genomic, transcriptomic, and imaging data. With this biobank they identify risk factors associated with progression of ductal carcinoma in situ to invasive breast cancer and create a collection of 19 distributable DCIS models, providing a resource for further research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
218. RAD51 testing in patients with early HER2-negative breast cancer and homologous recombination deficiency: post-hoc analysis of the GeparOla trial.
- Author
-
Villacampa G, Llop-Guevara A, Filmann N, Herencia A, Fasching PA, Karn T, Marmé F, Klare P, Müller V, Stefek A, Schem C, Uleer C, Fehm T, Doering G, Stickeler E, van Mackelenbergh M, Felder B, Nekljudova V, Balmaña J, Denkert C, Loibl S, and Serra V
- Abstract
Purpose: The randomized GeparOla trial reported comparable pathological complete response (pCR) rates with neoadjuvant containing olaparib vs. carboplatin treatment. Here, we evaluate the association between functional homologous repair deficiency (HRD) by RAD51 foci and pCR, and the potential of improving patient selection by combining RAD51 and stromal tumor infiltrating lymphocytes (sTILs)., Patients and Methods: This is a post-hoc blinded, biomarker analysis from the randomized GeparOla trial. Patients with early-stage HER2-negative breast cancer and HRD assessed by Myriad myChoice or BRCA1/BRCA2 mutation were randomized 1:1 to receive i) paclitaxel plus olaparib or ii) paclitaxel plus carboplatin, both followed by epirubicin/cyclophosphamide. Functional HRD was predefined as a RAD51 score ≤10% (RAD51-low)., Results: Overall, 90/97 (92.8%) samples were evaluable for RAD51 testing and 72/90 (80.0%) were RAD51-low. The pCR rate in patients with RAD51-low tumors was 66.7% (48/72), while it decreased to 22.2% (4/18) in those with RAD51-high. In the multivariable model including clinicopathological factors and treatment, the RAD51 score remained significantly associated with pCR (OR=12.03, 95%CI 2.60-55.73, p=0.002). Patients with RAD51-low and high sTILs in their tumors achieved a pCR rate of 75.0% (27/36). Similar results were observed for olaparib or carboplatin. In the exploratory DFS analysis, no differences were observed between RAD51 groups (high vs. low: HR=0.85, 95% CI 0.25-2.97)., Conclusions: In a pre-selected population with HRD according to a genetic test, RAD51 testing identifies patients with different pCR rates under PARPi or platinum-based therapies. Future biomarker-driven studies should consider this information to refine stratification factors and to improve patient selection.
- Published
- 2024
- Full Text
- View/download PDF
219. The PARP1 selective inhibitor saruparib (AZD5305) elicits potent and durable antitumor activity in patient-derived BRCA1/2-associated cancer models.
- Author
-
Herencia-Ropero A, Llop-Guevara A, Staniszewska AD, Domènech-Vivó J, García-Galea E, Moles-Fernández A, Pedretti F, Domènech H, Rodríguez O, Guzmán M, Arenas EJ, Verdaguer H, Calero-Nieto FJ, Talbot S, Tobalina L, Leo E, Lau A, Nuciforo P, Dienstmann R, Macarulla T, Arribas J, Díez O, Gutiérrez-Enríquez S, Forment JV, O'Connor MJ, Albertella M, Balmaña J, and Serra V
- Subjects
- Humans, Animals, Mice, Female, Phthalazines pharmacology, Phthalazines therapeutic use, Poly (ADP-Ribose) Polymerase-1 antagonists & inhibitors, Piperazines pharmacology, Piperazines therapeutic use, Indoles therapeutic use, Indoles pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Carboplatin pharmacology, Carboplatin therapeutic use, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, BRCA1 Protein genetics, BRCA2 Protein genetics, Xenograft Model Antitumor Assays
- Abstract
Background: Poly (ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors (PARPi) are targeted therapies approved for homologous recombination repair (HRR)-deficient breast, ovarian, pancreatic, and prostate cancers. Since inhibition of PARP1 is sufficient to cause synthetic lethality in tumors with homologous recombination deficiency (HRD), PARP1 selective inhibitors such as saruparib (AZD5305) are being developed. It is expected that selective PARP1 inhibition leads to a safer profile that facilitates its combination with other DNA damage repair inhibitors. Here, we aimed to characterize the antitumor activity of AZD5305 in patient-derived preclinical models compared to the first-generation PARP1/2 inhibitor olaparib and to identify mechanisms of resistance., Methods: Thirteen previously characterized patient-derived tumor xenograft (PDX) models from breast, ovarian, and pancreatic cancer patients harboring germline pathogenic alterations in BRCA1, BRCA2, or PALB2 were used to evaluate the efficacy of AZD5305 alone or in combination with carboplatin or an ataxia telangiectasia and Rad3 related (ATR) inhibitor (ceralasertib) and compared it to the first-generation PARPi olaparib. We performed DNA and RNA sequencing as well as protein-based assays to identify mechanisms of acquired resistance to either PARPi., Results: AZD5305 showed superior antitumor activity than the first-generation PARPi in terms of preclinical complete response rate (75% vs. 37%). The median preclinical progression-free survival was significantly longer in the AZD5305-treated group compared to the olaparib-treated group (> 386 days vs. 90 days). Mechanistically, AZD5305 induced more replication stress and genomic instability than the PARP1/2 inhibitor olaparib in PARPi-sensitive tumors. All tumors at progression with either PARPi (39/39) showed increase of HRR functionality by RAD51 foci formation. The most prevalent resistance mechanisms identified were the acquisition of reversion mutations in BRCA1/BRCA2 and the accumulation of hypomorphic BRCA1. AZD5305 did not sensitize PDXs with acquired resistance to olaparib but elicited profound and durable responses when combined with carboplatin or ceralasertib in 3/6 and 5/5 models, respectively., Conclusions: Collectively, these results show that the novel PARP1 selective inhibitor AZD5305 yields a potent antitumor response in PDX models with HRD and delays PARPi resistance alone or in combination with carboplatin or ceralasertib, which supports its use in the clinic as a new therapeutic option., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
220. Functional HRD by RAD51 identifies BRCA1 VUS associated with loss of gene function and response to DNA-damaging agents.
- Author
-
Casartelli C, Tommasi C, Lazzarin A, Corianò M, Tornali C, Serra O, Campanini N, Gutiérrez-Enríquez S, Sikokis A, Zanoni D, Minari R, Bortesi B, Michiara M, Boggiani D, Uliana V, Llop-Guevara A, Serra V, Musolino A, and Pellegrino B
- Subjects
- Humans, Female, Loss of Function Mutation, Breast Neoplasms genetics, Breast Neoplasms drug therapy, BRCA1 Protein metabolism, BRCA1 Protein genetics, Rad51 Recombinase metabolism, DNA Damage
- Published
- 2024
- Full Text
- View/download PDF
221. Longitudinal profiling identifies co-occurring BRCA1/2 reversions, TP53BP1, RIF1 and PAXIP1 mutations in PARP inhibitor-resistant advanced breast cancer.
- Author
-
Harvey-Jones E, Raghunandan M, Robbez-Masson L, Magraner-Pardo L, Alaguthurai T, Yablonovitch A, Yen J, Xiao H, Brough R, Frankum J, Song F, Yeung J, Savy T, Gulati A, Alexander J, Kemp H, Starling C, Konde A, Marlow R, Cheang M, Proszek P, Hubank M, Cai M, Trendell J, Lu R, Liccardo R, Ravindran N, Llop-Guevara A, Rodriguez O, Balmana J, Lukashchuk N, Dorschner M, Drusbosky L, Roxanis I, Serra V, Haider S, Pettitt SJ, Lord CJ, and Tutt ANJ
- Subjects
- Female, Humans, BRCA1 Protein genetics, BRCA2 Protein genetics, Homologous Recombination, Mutation, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Tumor Suppressor p53-Binding Protein 1, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Resistance to therapies that target homologous recombination deficiency (HRD) in breast cancer limits their overall effectiveness. Multiple, preclinically validated, mechanisms of resistance have been proposed, but their existence and relative frequency in clinical disease are unclear, as is how to target resistance., Patients and Methods: Longitudinal mutation and methylation profiling of circulating tumour (ct)DNA was carried out in 47 patients with metastatic BRCA1-, BRCA2- or PALB2-mutant breast cancer treated with HRD-targeted therapy who developed progressive disease-18 patients had primary resistance and 29 exhibited response followed by resistance. ctDNA isolated at multiple time points in the patient treatment course (before, on-treatment and at progression) was sequenced using a novel >750-gene intron/exon targeted sequencing panel. Where available, matched tumour biopsies were whole exome and RNA sequenced and also used to assess nuclear RAD51., Results: BRCA1/2 reversion mutations were present in 60% of patients and were the most prevalent form of resistance. In 10 cases, reversions were detected in ctDNA before clinical progression. Two new reversion-based mechanisms were identified: (i) intragenic BRCA1/2 deletions with intronic breakpoints; and (ii) intragenic BRCA1/2 secondary mutations that formed novel splice acceptor sites, the latter being confirmed by in vitro minigene reporter assays. When seen before commencing subsequent treatment, reversions were associated with significantly shorter time to progression. Tumours with reversions retained HRD mutational signatures but had functional homologous recombination based on RAD51 status. Although less frequent than reversions, nonreversion mechanisms [loss-of-function (LoF) mutations in TP53BP1, RIF1 or PAXIP1] were evident in patients with acquired resistance and occasionally coexisted with reversions, challenging the notion that singular resistance mechanisms emerge in each patient., Conclusions: These observations map the prevalence of candidate drivers of resistance across time in a clinical setting, information with implications for clinical management and trial design in HRD breast cancers., Competing Interests: Disclosure ANJT is/has been a consultant for AstraZeneca, Merck KGaA, Artios, Pfizer, Vertex, GE Healthcare, Inbiomotion, Prime Oncology, Medscape Education, EMPartners, VJ Oncoclogy, Gilead and MD Anderson Cancer Centre; has received grant/research support from AstraZeneca, Myriad, Medivation and Merck KGaA; is a stockholder in Inbiomotion; is also a named inventor on patents describing the use of DNA repair inhibitors and stands to gain from their development and use as part of the ICR ‘Rewards to Inventors’ scheme and also reports benefits from this scheme associated with patents for PARP inhibitors paid to ANJT research accounts at the Institute of Cancer Research. CJL receives and/or has received research funding from AstraZeneca, Merck KGaA and Artios; received consultancy, SAB membership or honoraria payments from Syncona, Sun Pharma, Gerson Lehrman Group, Merck KGaA, Vertex, AstraZeneca, Tango, 3rd Rock, Ono Pharma, Artios, Abingworth, Tesselate, Dark Blue Therapeutics, Pontifax, Astex, Neophore, Glaxo Smith Kline; has stock in Tango, Ovibio, Hysplex and Tesselate; is also a named inventor on patents describing the use of DNA repair inhibitors and stands to gain from their development and use as part of the ICR ‘Rewards to Inventors’ scheme and also reports benefits from this scheme associated with patents for PARP inhibitors paid into CJL’s personal account and research accounts at the Institute of Cancer Research. AY, JY, MD, and LD are employees and stockholders of Guardant Health. JY is a former full-time employee at Guardant Health and a current full-time employee at Exai Bio. SJP is a named inventor on patents relating to targeting PARPi resistance and stands to gain from their development and use as part of the ICR ‘Rewards to Inventors’ scheme. VS has research grants and receives honoraria from AstraZeneca. ALG and VS are co-inventors of the patent PCT/EP2018/086759 (WO2019122411A1). VS and ALG received funding from Fundació La Marató de TV3 (654/C/2019), FERO-GHD and AECC (INVES20095LLOP). NL is employed by AstraZeneca and owns AstraZeneca stock. The other authors declare no conflict of interest., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
222. Olaparib and Ceralasertib (AZD6738) in Patients with Triple-Negative Advanced Breast Cancer: Results from Cohort E of the plasmaMATCH Trial (CRUK/15/010).
- Author
-
Ring A, Kilburn LS, Pearson A, Moretti L, Afshari-Mehr A, Wardley AM, Gurel B, Macpherson IR, Riisnaes R, Baird RD, Martin S, Roylance R, Johnson H, Ferreira A, Winter MC, Dunne K, Copson E, Hickish T, Burcombe R, Randle K, Serra V, Llop-Guevara A, Bliss JM, and Turner NC
- Subjects
- Humans, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, BRCA1 Protein genetics, BRCA2 Protein genetics, Phthalazines adverse effects, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Antineoplastic Agents therapeutic use
- Abstract
Purpose: Approximately 10% to 15% of triple-negative breast cancers (TNBC) have deleterious mutations in BRCA1 and BRCA2 and may benefit from PARP inhibitor treatment. PARP inhibitors may also increase exogenous replication stress and thereby increase sensitivity to inhibitors of ataxia telangiectasia and Rad3-related (ATR) protein. This phase II study examined the activity of the combination of PARP inhibitor, olaparib, and ATR inhibitor, ceralasertib (AZD6738), in patients with advanced TNBC., Patients and Methods: Patients with TNBC on most recent biopsy who had received 1 or 2 lines of chemotherapy for advanced disease or had relapsed within 12 months of (neo)adjuvant chemotherapy were eligible. Treatment was olaparib 300 mg twice a day continuously and celarasertib 160 mg on days 1-7 on a 28-day cycle until disease progression. The primary endpoint was confirmed objective response rate (ORR). Tissue and plasma biomarker analyses were preplanned to identify predictors of response., Results: 70 evaluable patients were enrolled. Germline BRCA1/2 mutations were present in 10 (14%) patients and 3 (4%) patients had somatic BRCA mutations. The confirmed ORR was 12/70; 17.1% (95% confidence interval, 10.4-25.5). Responses were observed in patients without germline or somatic BRCA1/2 mutations, including patients with mutations in other homologous recombination repair genes and tumors with functional homologous recombination deficiency by RAD51 foci., Conclusions: The response rate to olaparib and ceralasertib did not meet prespecified criteria for activity in the overall evaluable population, but responses were observed in patients who would not be expected to respond to olaparib monotherapy., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
223. RAD51 Foci as a Biomarker Predictive of Platinum Chemotherapy Response in Ovarian Cancer.
- Author
-
Compadre AJ, van Biljon LN, Valentine MC, Llop-Guevara A, Graham E, Fashemi B, Herencia-Ropero A, Kotnik EN, Cooper I, Harrington SP, Kuroki LM, McCourt CK, Hagemann AR, Thaker PH, Mutch DG, Powell MA, Sun L, Mosammaparast N, Serra V, Zhao P, Lomonosova E, Khabele D, and Mullen MM
- Subjects
- Humans, Female, Carcinoma, Ovarian Epithelial drug therapy, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Biomarkers, Tumor therapeutic use, Platinum therapeutic use, Ovarian Neoplasms pathology
- Abstract
Purpose: To determine the ability of RAD51 foci to predict platinum chemotherapy response in high-grade serous ovarian cancer (HGSOC) patient-derived samples., Experimental Design: RAD51 and γH2AX nuclear foci were evaluated by immunofluorescence in HGSOC patient-derived cell lines (n = 5), organoids (n = 11), and formalin-fixed, paraffin-embedded tumor samples (discovery n = 31, validation n = 148). Samples were defined as RAD51-High if >10% of geminin-positive cells had ≥5 RAD51 foci. Associations between RAD51 scores, platinum chemotherapy response, and survival were evaluated., Results: RAD51 scores correlated with in vitro response to platinum chemotherapy in established and primary ovarian cancer cell lines (Pearson r = 0.96, P = 0.01). Organoids from platinum-nonresponsive tumors had significantly higher RAD51 scores than those from platinum-responsive tumors (P < 0.001). In a discovery cohort, RAD51-Low tumors were more likely to have a pathologic complete response (RR, 5.28; P < 0.001) and to be platinum-sensitive (RR, ∞; P = 0.05). The RAD51 score was predictive of chemotherapy response score [AUC, 0.90; 95% confidence interval (CI), 0.78-1.0; P < 0.001). A novel automatic quantification system accurately reflected the manual assay (92%). In a validation cohort, RAD51-Low tumors were more likely to be platinum-sensitive (RR, ∞; P < 0.001) than RAD51-High tumors. Moreover, RAD51-Low status predicted platinum sensitivity with 100% positive predictive value and was associated with better progression-free (HR, 0.53; 95% CI, 0.33-0.85; P < 0.001) and overall survival (HR, 0.43; 95% CI, 0.25-0.75; P = 0.003) than RAD51-High status., Conclusions: RAD51 foci are a robust marker of platinum chemotherapy response and survival in ovarian cancer. The utility of RAD51 foci as a predictive biomarker for HGSOC should be tested in clinical trials., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
224. BRCA2 Germline Mutations Identify Gastric Cancers Responsive to PARP Inhibitors.
- Author
-
Petrelli A, Rizzolio S, Pietrantonio F, Bellomo SE, Benelli M, De Cecco L, Romagnoli D, Berrino E, Orrù C, Ribisi S, Moya-Rull D, Migliore C, Conticelli D, Maina IM, Puliga E, Serra V, Pellegrino B, Llop-Guevara A, Musolino A, Siena S, Sartore-Bianchi A, Prisciandaro M, Morano F, Antista M, Fumagalli U, De Manzoni G, Degiuli M, Baiocchi GL, Amisano MF, Ferrero A, Marchiò C, Corso S, and Giordano S
- Subjects
- Humans, Female, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Germ-Line Mutation, Retrospective Studies, BRCA1 Protein genetics, BRCA2 Protein genetics, Stomach Neoplasms drug therapy, Stomach Neoplasms genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Ovarian Neoplasms drug therapy
- Abstract
Despite negative results of clinical trials conducted on the overall population of patients with gastric cancer, PARP inhibitor (PARPi) therapeutic strategy still might represent a window of opportunity for a subpopulation of patients with gastric cancer. An estimated 7% to 12% of gastric cancers exhibit a mutational signature associated with homologous recombination (HR) failure, suggesting that these patients could potentially benefit from PARPis. To analyze responsiveness of gastric cancer to PARPi, we exploited a gastroesophageal adenocarcinoma (GEA) platform of patient-derived xenografts (PDX) and PDX-derived primary cells and selected 10 PDXs with loss-of-function mutations in HR pathway genes. Cell viability assays and preclinical trials showed that olaparib treatment was effective in PDXs harboring BRCA2 germline mutations and somatic inactivation of the second allele. Olaparib responsive tumors were sensitive to oxaliplatin as well. Evaluation of HR deficiency (HRD) and mutational signatures efficiently stratified responder and nonresponder PDXs. A retrospective analysis on 57 patients with GEA showed that BRCA2 inactivating variants were associated with longer progression-free survival upon platinum-based regimens. Five of 7 patients with BRCA2 germline mutations carried the p.K3326* variant, classified as "benign." However, familial history of cancer, the absence of RAD51 foci in tumor cells, and a high HRD score suggest a deleterious effect of this mutation in gastric cancer. In conclusion, PARPis could represent an effective therapeutic option for BRCA2-mutated and/or high HRD score patients with GEA, including patients with familial intestinal gastric cancer., Significance: PARP inhibition is a potential strategy for treating patients with gastric cancer with mutated BRCA2 or homologous repair deficiency, including patients with familial intestinal gastric cancer, for whom BRCA2 germline testing should be recommended., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
225. Human Metastatic Cholangiocarcinoma Patient-Derived Xenografts and Tumoroids for Preclinical Drug Evaluation.
- Author
-
Serra-Camprubí Q, Verdaguer H, Oliveros W, Lupión-Garcia N, Llop-Guevara A, Molina C, Vila-Casadesús M, Turpin A, Neuzillet C, Frigola J, Querol J, Yáñez-Bartolomé M, Castet F, Fabregat-Franco C, Escudero-Iriarte C, Escorihuela M, Arenas EJ, Bernadó-Morales C, Haro N, Giles FJ, Pozo ÓJ, Miquel JM, Nuciforo PG, Vivancos A, Melé M, Serra V, Arribas J, Tabernero J, Peiró S, Macarulla T, and Tian TV
- Subjects
- Humans, Drug Evaluation, Preclinical, Heterografts, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Bile Ducts, Intrahepatic, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics
- Abstract
Purpose: Cholangiocarcinoma (CCA) is usually diagnosed at advanced stages, with limited therapeutic options. Preclinical models focused on unresectable metastatic CCA are necessary to develop rational treatments. Pathogenic mutations in IDH1/2, ARID1A/B, BAP1, and BRCA1/2 have been identified in 30%-50% of patients with CCA. Several types of tumor cells harboring these mutations exhibit homologous recombination deficiency (HRD) phenotype with enhanced sensitivity to PARP inhibitors (PARPi). However, PARPi treatment has not yet been tested for effectiveness in patient-derived models of advanced CCA., Experimental Design: We have established a collection of patient-derived xenografts from patients with unresectable metastatic CCA (CCA_PDX). The CCA_PDXs were characterized at both histopathologic and genomic levels. We optimized a protocol to generate CCA tumoroids from CCA_PDXs. We tested the effects of PARPis in both CCA tumoroids and CCA_PDXs. Finally, we used the RAD51 assay to evaluate the HRD status of CCA tissues., Results: This collection of CCA_PDXs recapitulates the histopathologic and molecular features of their original tumors. PARPi treatments inhibited the growth of CCA tumoroids and CCA_PDXs with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1. In line with these findings, only CCA_PDX and CCA patient biopsy samples with mutations of BRCA2 showed RAD51 scores compatible with HRD., Conclusions: Our results suggest that patients with advanced CCA with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1, are likely to benefit from PARPi therapy. This collection of CCA_PDXs provides new opportunities for evaluating drug response and prioritizing clinical trials., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
226. Alternative academic approaches for testing homologous recombination deficiency in ovarian cancer in the MITO16A/MaNGO-OV2 trial.
- Author
-
Capoluongo ED, Pellegrino B, Arenare L, Califano D, Scambia G, Beltrame L, Serra V, Scaglione GL, Spina A, Cecere SC, De Cecio R, Normanno N, Colombo N, Lorusso D, Russo D, Nardelli C, D'Incalci M, Llop-Guevara A, Pisano C, Baldassarre G, Mezzanzanica D, Artioli G, Setaro M, Tasca G, Roma C, Campanini N, Cinieri S, Sergi A, Musolino A, Perrone F, Chiodini P, Marchini S, and Pignata S
- Subjects
- Female, Humans, Bevacizumab therapeutic use, Carboplatin therapeutic use, Homologous Recombination, Paclitaxel therapeutic use, Platinum therapeutic use, Poly(ADP-ribose) Polymerases genetics, Poly(ADP-ribose) Polymerases therapeutic use, Mangifera, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics
- Abstract
Background: The detection of homologous recombination deficiency (HRD) can identify patients who are more responsive to platinum and poly ADP ribose polymerase inhibitors (PARPi). MyChoice CDx (Myriad) is the most used HRD test in ovarian cancer (OC). However, some limitations of commercial tests exist, because of the high rate of inconclusive results, costs, and the impossibility of evaluating functional resistance mechanisms., Patients and Methods: Two academic genomic tests and a functional assay, the RAD51 foci, were evaluated to detect HRD. One hundred patients with high-grade OC enrolled in the MITO16A/MaNGO-OV2 trial and treated with first-line therapy with carboplatin, paclitaxel, and bevacizumab were analyzed., Results: The failure rate of the two genomic assays was 2%. The sensitivity in detecting HRD when compared with Myriad was 98.1% and 90.6%, respectively. The agreement rate with Myriad was 0.92 and 0.87, with a Cohen's κ coefficient corresponding to 0.84 and 0.74, respectively. For the RAD51 foci assay, the failure rate was 30%. When the test was successful, discordant results for deficient and proficient tumors were observed, and additional HRD patients were identified compared to Myriad; sensitivity was 82.9%, agreement rate was 0.65, and Cohen's κ coefficient was 0.18. The HRD detected by genomic assays and residual tumor at primary surgery and stage was correlated with progression-free survival at multivariate analysis., Conclusions: Results suggest the feasibility of academic tests for assessing HRD status that show robust concordance with Myriad and correlation with clinical outcome. The contribution of the functional information related to the RAD51 foci test to the genomic data needs further investigation., (Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
227. Association of RAD51 with homologous recombination deficiency (HRD) and clinical outcomes in untreated triple-negative breast cancer (TNBC): analysis of the GeparSixto randomized clinical trial.
- Author
-
Llop-Guevara A, Loibl S, Villacampa G, Vladimirova V, Schneeweiss A, Karn T, Zahm DM, Herencia-Ropero A, Jank P, van Mackelenbergh M, Fasching PA, Marmé F, Stickeler E, Schem C, Dienstmann R, Florian S, Nekljudova V, Balmaña J, Hahnen E, Denkert C, and Serra V
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, BRCA1 Protein genetics, Carboplatin therapeutic use, Homologous Recombination, Humans, Rad51 Recombinase genetics, Randomized Controlled Trials as Topic, Retrospective Studies, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics
- Abstract
Background: Current genetic and genomic tests measuring homologous recombination deficiency (HRD) show limited predictive value. This study compares the performance of an immunohistology-based RAD51 test with genetic/genomic tests to identify patients with HRD primary triple-negative breast cancer (TNBC) and evaluates its accuracy to select patients sensitive to platinum-based neoadjuvant chemotherapy (NACT)., Patients and Methods: This is a retrospective, blinded, biomarker analysis from the GeparSixto randomized clinical trial. TNBC patients received neoadjuvant paclitaxel plus Myocet®-nonpegylated liposomal doxorubicin (PM) or PM plus carboplatin (PMCb), both arms including bevacizumab. Formalin-fixed paraffin-embedded (FFPE) tumor samples were laid on tissue microarrays. RAD51, BRCA1 and γH2AX were quantified using an immunofluorescence assay. The predictive value of RAD51 was assessed by regression models. Concordance analyses were carried out between RAD51 score and tumor BRCA (tBRCA) status or genomic HRD score (Myriad myChoice®). Associations with pathological complete response (pCR) and survival were studied. Functional HRD was predefined as a RAD51 score ≤10% (RAD51-low)., Results: Functional HRD by RAD51-low was evidenced in 81/133 tumors (61%). RAD51 identified 93% tBRCA-mutated tumors and 45% non-tBRCA mutant cases as functional HRD. The concordance between RAD51 and genomic HRD was 87% [95% confidence interval (CI) 79% to 93%]. In patients with RAD51-high tumors, pCR was similar between treatment arms [PMCb 31% versus PM 39%, odds ratio (OR) 0.71, 0.23-2.24, P = 0.56]. Patients with RAD51-low tumors benefited from PMCb (pCR 66% versus 33%, OR 3.96, 1.56-10.05, P = 0.004; interaction test P = 0.02). This benefit maintained statistical significance in the multivariate analysis. Carboplatin addition showed similar disease-free survival in the RAD51-high [hazard ratio (HR) 0.40, log-rank P = 0.11] and RAD51-low (0.45, P = 0.11) groups., Conclusions: The RAD51 test identifies tumors with functional HRD and is highly concordant with tBRCA mutation and genomic HRD. RAD51 independently predicts clinical benefit from adding Cb to NACT in TNBC. Our results support further development to incorporate RAD51 testing in clinical decision-making., Competing Interests: Disclosure Personal fees and/or nonfinancial support from: Agendia (FM), Amgen (RD, MvM), AstraZeneca (JB, CD, MvM), Boehringer Ingelheim (RD), Celgene (FM), Chugai (SL), Clovis (FM), Daiichi-Sankyo (SL, CD), Genomic Health (MvM), GSK (FM), Immunomedics (FM), Ipsen (RD), Janssen-Cilag (FM), Libbs (RD), Lilly (FM), Merck-MSD (RD, FM, CD), Molecular Health (CD), Mylan (MvM), Novartis (FM, CD, MvM), Pfizer (FM, JB, MvM), Pierre Fabre (FM, MvM), Roche (RD, CD, MvM), Sanofi (RD), SeaGen (FM), Servier (RD). Research grants from: Abbvie (SL), Amgen (SL), AstraZeneca (SL, VS), Celgene (SL), Daiichi-Sankyo (SL), Merck (RD), Myriad Genetics (PJ, CD), Novartis (SL), Pierre Fabre (RD), Pfizer (SL), Roche (SL, CD), Tesaro (VS), Teva (SL), Vifor (SL), Immunomedics (SL). Other funding paid to institution from: BMS (SL), Eirgenix (SL), Lilly (SL), Merck (SL), MSD (SL), SeaGen (SL), Prime/Medscape (SL), Puma (SL), Samsung (SL), Pierre Fabre (SL). Stock ownership from: Myriad Genetics (PJ), Sividon Diagnostics (CD until 2016). Patents and other intellectual property: WO2019122411A1 (ALG, JB, VS), EP14153692.0 (SL), VMScope digital pathology software (CD), WO2015114146A1 (CD), WO2010076322A1 (CD), WO2020109570A1 (CD). All remaining authors have declared no conflicts of interest., (Copyright © 2021 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.