bims-ovdlit Biomed News
on Ovarian cancer: early diagnosis, liquid biopsy and therapy
Issue of 2021‒12‒19
twelve papers selected by
Lara Paracchini
Humanitas Research


  1. Gynecol Oncol. 2021 Dec 11. pii: S0090-8258(21)01630-9. [Epub ahead of print]
      OBJECTIVE: In the Phase 3 VELIA trial (NCT02470585), PARP inhibitor (PARPi) veliparib was combined with first-line chemotherapy and continued as maintenance for patients with ovarian carcinoma enrolled regardless of chemotherapy response or biomarker status. Here, we report exploratory analyses of the impact of homologous recombination deficient (HRD) or proficient (HRP) status on progression-free survival (PFS) and objective response rates during chemotherapy.METHODS: Women with Stage III-IV ovarian carcinoma were randomized to veliparib-throughout, veliparib-combination-only, or placebo. Stratification factors included timing of surgery and germline BRCA mutation status. HRD status was dichotomized at genomic instability score 33. During combination therapy, CA-125 levels were measured at baseline and each cycle; radiographic responses were assessed every 9 weeks.
    RESULTS: Of 1140 patients randomized, 742 had BRCA wild type (BRCAwt) tumors (HRP, n = 373; HRD/BRCAwt, n = 329). PFS hazard ratios between veliparib-throughout versus control were similar in both BRCAwt populations (HRD/BRCAwt: 22.9 vs 19.8 months; hazard ratio 0.76; 95% confidence interval [CI] 0.53-1.09; HRP: 15.0 vs 11.5 months; hazard ratio 0.765; 95% CI 0.56-1.04). By Cycle 3, the proportion with ≥90% CA-125 reduction from baseline was higher in those receiving veliparib (pooled arms) versus control (34% vs 23%; P = 0.0004); particularly in BRCAwt and HRP subgroups. Complete response rates among patients with measurable disease after surgery were 24% with veliparib (pooled arms) and 18% with control.
    CONCLUSIONS: These results potentially broaden opportunities for PARPi utilization among patients who would not qualify for frontline PARPi maintenance based on other trials.
    Keywords:  BRCA1/2; Homologous recombination deficiency; Veliparib
    DOI:  https://doi.org/10.1016/j.ygyno.2021.12.003
  2. Nature. 2021 Dec;600(7889): S40-S41
      
    Keywords:  Cancer; Microbiome
    DOI:  https://doi.org/10.1038/d41586-021-03716-8
  3. Nature. 2021 Dec;600(7889): S48-S49
      
    Keywords:  Cancer; Epidemiology
    DOI:  https://doi.org/10.1038/d41586-021-03719-5
  4. Nature. 2021 Dec;600(7889): S35
      
    Keywords:  Cancer; Health care; Therapeutics
    DOI:  https://doi.org/10.1038/d41586-021-03713-x
  5. Nature. 2021 Dec;600(7889): S42-S44
      
    Keywords:  Cancer; Health care
    DOI:  https://doi.org/10.1038/d41586-021-03717-7
  6. Cancer J. 2021 Nov-Dec 01;27(6):27(6): 511-520
      ABSTRACT: The use of poly(ADP-ribose) polymerase inhibitors and immune checkpoint inhibitor therapies has seen substantial clinical success in oncology therapeutic development. Although multiple agents within these classes have achieved regulatory approval globally-in several malignancies in early and advanced stages-drug resistance remains an issue. Building on preclinical evidence, several early trials and late-phase studies are underway. This review explores the therapeutic potential of combination poly(ADP-ribose) polymerase inhibitors and immune checkpoint inhibitor therapy in solid tumors, including the scientific and therapeutic rationale, available clinical evidence, and considerations for future trial and biomarker development across different malignancies using ovarian and other solid cancer subtypes as key examples.
    DOI:  https://doi.org/10.1097/PPO.0000000000000557
  7. Methods Mol Biol. 2022 ;2424 135-146
      Cancer is a complex disease rooted in heterogeneity, which is the phenomenon of individual cells, tissues, or patients having distinct phenotypic and/or genetic characteristics. Observed divergent disease etiology is likely rooted, at least in part, in tumor heterogeneity and the classification of distinct and important subpopulations of cells within the tumor and its associated microenvironment has remained a technical challenge. Standard next-generation sequencing of bulk tumor tissue provides an overall average genetic profile of the sample, and masks contributions from individual cells and minor populations of cells, particularly in heterogeneous samples. Only with the advent of single-cell analysis and sequencing technologies has it become possible to characterize key contributions of cellular subpopulations in order to more comprehensively characterize disease. This chapter describes a method to generate linked phenotypic and genotypic data at single-cell resolution using a real-time single-cell resolved platform. Specifically, the example method provided here is used to link cellular growth kinetics and expression of a prognostic marker protein, CA-125, in cells derived from ovarian cancer patients with their single-cell genomic profiles, but the method is translatable to other cell types and phenotypes of interest.
    Keywords:  Ovarian cancer; Single-cell DNA Seq; Single-cell RNA Seq; Single-cell analysis; Tumor heterogeneity
    DOI:  https://doi.org/10.1007/978-1-0716-1956-8_8
  8. Nature. 2021 Dec;600(7889): S45-S47
      
    Keywords:  Cancer; Health care; Therapeutics
    DOI:  https://doi.org/10.1038/d41586-021-03718-6
  9. Nature. 2021 Dec;600(7889): S39
      
    Keywords:  Cancer; Health care; Society
    DOI:  https://doi.org/10.1038/d41586-021-03715-9
  10. Nat Commun. 2021 Dec 16. 12(1): 7353
      
    DOI:  https://doi.org/10.1038/s41467-021-27389-z
  11. Nature. 2021 Dec;600(7889): S53
      
    Keywords:  Cancer; Medical research
    DOI:  https://doi.org/10.1038/d41586-021-03721-x