20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The DNA damage repair (DDR) pathway in biliary tract cancer (BTC): a new Pandora’s box?

      letter
      , ,
      ESMO Open
      BMJ Publishing Group
      correspondence

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          We read with great interest the article ‘Molecular profile of BRCA-mutated biliary tract cancers’ by Spizzo and colleagues.1 This study provides substantial evidence of the emerging importance of BRCA 1/2 mutations (BRCAm) identification in biliary tract cancers (BTC), suggesting a potential association between BRCAm and response to immune checkpoint inhibitors (ICIs). The emerging role of these genomic alterations as novel therapeutic target in metastatic solid tumours has led to enhance the concept of patient-focused, personalised medicine, paving the way towards new therapeutic scenarios in poor-prognosis malignancies. For this reason, the results reported by Spizzo and colleagues add another significant element to implement tailor-made treatment strategies for patients with BTC. However, some questions remain open. First, the prevalence of BRCAm fluctuates from 1% to 7% across BTC, while a larger spectrum of genes which compromise DNA damage repair (DDR) pathway have been reported to occur in up to 28.9% patients with newly diagnosed BTC.2 Interestingly, detection of multiple loss-of-function mutations in other DDR genes, epigenetic inactivation of BRCA1 or methylation of RAD51C promoters may identify candidates for displaying ‘BRCAness’ phenotype.2 Nevertheless, there is still no consensus on methods for testing and defining DDR alterations in BTC. A recent study by Park and colleagues3 proposed a selection of 17 germline and somatic alteration of HRR genes (ATM, BAP1, BARD1, BLM, BRCA1, BRCA2, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, PALB2, RAD50, RAD51, RAD51C and RTEL1) in addition to BRCAm to evaluate a correlation with genomic instability in patients affected by pancreatic ductal adenocarcinoma. Moreover, despite the exciting therapeutic potential of DNA-damaging agents in these patients with broader evidence of ‘BRCAness’, how DDR pathway deficiency could associate with clinical responses and patient outcomes in BTC is still unknown. Another question may deserve attention. Two months ago, Kim and colleagues4 published results from a multicentre, phase II trial evaluating nivolumab in 54 patients with BTC after progression on 1 up to 3 lines of systemic therapy. According to the results of this study, 22% (10/54) of patients receiving nivolumab achieved objective response per RECIST V.1.1 criteria and, surprisingly, all responders had a mismatch-repair proficient tumour.4 Therefore, considering the recent results reported by Spizzo et al and the approximate value of prevalence of DDR gene mutations in BTC, it would be interesting to know how many patients with DDR alterations were included in the study and if a proportion of ICIs responders harboured a ‘BRCAness’ phenotype. Recently, comprehensive genomic analysis of 198 advanced non-small-cell lung cancer (NSCLC) samples carried out by Shim and colleagues5 suggested that homologous recombination deficiency (HRD) could influence the response to ICIs by activating the stimulator of interferon genes signalling pathway and HRD could be associated with higher tumour mutation burden and longer progression-free survival in NSCLC. These findings provide an intriguing speculation regarding the potential therapeutic implications of DDR alterations across solid tumours. In summary, we believe further efforts are needed to ascertain DDR deficiency in a comprehensive and cost-effective approach and to explore the potential role of DDR alterations as potential predictive biomarker in BTC.

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: not found

          A Phase 2 Multi-institutional Study of Nivolumab for Patients With Advanced Refractory Biliary Tract Cancer

          Currently, there is no established second-line systemic treatment for biliary tract cancer (BTC). Preclinical data have demonstrated that the presence of tumor-infiltrating CD8 T cells and programmed cell death 1 ligand 1-expressing tumor cells in the tumor microenvironment of BTC supports the rationale of using programmed cell death 1 protein blockade immunotherapy in BTC.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Prevalence of Homologous Recombination–Related Gene Mutations Across Multiple Cancer Types

            The prevalence of homologous recombination DNA damage repair (HR-DDR) deficiencies among all tumor lineages is not well characterized. Therapy directed toward homologous recombination DDR deficiency (HRD) is now approved in ovarian and breast cancer, and there may be additional opportunities for benefit for patients with other cancers. Comprehensive evaluations for HRD are limited in part by the lack of a uniform, cost-effective method for testing and defining HRD. Molecular profiles of 52,426 tumors were reviewed to identify pathogenic mutations in the HR-DDR genes ARID1A, ATM, ATRX, BAP1, BARD1, BLM, BRCA1/2, BRIP1, CHEK1/2, FANCA/C/D2/E/F/G/L, MRE11A, NBN, PALB2, RAD50, RAD51, RAD51B, or WRN. From solid tumors submitted to Caris Life Sciences, molecular profiles were generated using next-generation sequencing (NGS; average read depth, 500×). A total of 17,566 tumors were sequenced with NGS600 (n = 592 genes), and 34,860 tumors underwent hotspot Illumina MiSeq platform testing (n = 47 genes). Of the tumors that underwent NGS600 testing, the overall frequency of HR-DDR mutations detected was 17.4%, and the most commonly mutated lineages were endometrial (34.4%; n = 1,475), biliary tract (28.9%; n = 343), bladder (23.9%; n = 201), hepatocellular (20.9%; n = 115), gastroesophageal (20.8%; n = 619), and ovarian (20.0%; n = 2,489). Least commonly mutated lineages included GI stromal (3.7%; n = 108), head and neck (6.8%; n = 206), and sarcoma (9.3%; n = 592). ARID1A was the most commonly mutated gene (7.2%), followed by BRCA2 (3.0%), BRCA1 (2.8%), ATM (1.3%), ATRX (1.3%), and CHEK2 (1.3%). HR-DDR mutations were seen in 17.4% of tumors across 21 cancer lineages, providing a path to explore the role of HRD-directed therapies, including poly-ADP ribose polymerase inhibitors, DNA-damaging chemotherapies, and newer agents such as ATR inhibitors.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection

              Genomic methods can identify homologous recombination deficiency (HRD). Rigorous evaluation of their outcome association to DNA-damage-response-targeted therapies like platinum in pancreatic cancer (PDAC) is essential in maximizing therapeutic outcome. We evaluated progression-free survival (PFS) and overall survival (OS) of advanced-stage PDAC patients, who had both germline and somatic targeted-gene sequencing. Homologous recombination-gene mutations (HRm) were evaluated. ( BRCA1, BRCA2, PALB2, ATM, BAP1, BARD1, BLM, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, RAD50, RAD51, RAD51C, RTEL1) HRm status was grouped as: (1) germline vs. somatic; (2) core ( BRCAs, PALB2) vs. non-core (other HRm); and (3) monoallelic vs. biallelic. Genomic instability was compared using large-scale state transition, signature 3, and tumor mutation burden. Among 262 patients, 50 (19%) had HRD (15% germline and 4% somatic). Both groups were analyzed together due to lack of difference in their genomic instability and outcome. Median [95%CI] follow-up was 21.9 [1.4–57.0] months. Median OS and PFS were 15.5 [14.6–19] and 7 [6.1–8.1] months, respectively. Patients with HRD had improved PFS compared to no HRD when treated with first-line (1L) platinum (HR: 0.44 [95%CI: 0.29–0.67], p <0.01), but not with 1L-non-platinum. Multivariable analysis showed HRD patients had improved OS regardless of their 1L-treatment, but most had platinum exposure during their course. Biallelic HRm (11%) and core HRm (12%) had higher genomic instability, which translated to improved PFS on 1L-platinum vs. 1L-non-platinum. Pathogenic HRm identifies HRD in PDAC patients with the best outcome when treated with 1L-platinum. Biallelic HRm and core HRm further enriched benefit from HRD.
                Bookmark

                Author and article information

                Journal
                ESMO Open
                ESMO Open
                esmoopen
                esmoopen
                ESMO Open
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                2059-7029
                2020
                29 September 2020
                : 5
                : 5
                : e001042
                Affiliations
                [1]departmentDepartment of Experimental, Diagnostic and Specialty Medicine , S. Orsola-Malpighi University Hospital , Bologna, Italy
                Author notes
                [Correspondence to ] Dr Angela Dalia Ricci; dalia.ricci@ 123456gmail.com
                Author information
                http://orcid.org/0000-0002-0701-6764
                Article
                esmoopen-2020-001042
                10.1136/esmoopen-2020-001042
                7526276
                32994319
                47bfd461-48a0-4db4-a0b0-71225ab28198
                © Author (s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. Published by BMJ on behalf of the European Society for Medical Oncology.

                This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, any changes made are indicated, and the use is non-commercial. See:  http://creativecommons.org/licenses/by-nc/4.0/.

                History
                : 12 September 2020
                : 12 September 2020
                Categories
                Correspondence
                1506
                Custom metadata
                unlocked

                correspondence
                correspondence

                Comments

                Comment on this article