8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Final results regarding the addition of dendritic cell vaccines to neoadjuvant chemotherapy in early HER2-negative breast cancer patients: clinical and translational analysis

      research-article

      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

          Background:

          Primary breast cancer (BC) has shown a higher immune infiltration than the metastatic disease, justifying the optimal scenario for immunotherapy. Recently, neoadjuvant chemotherapy (NAC) combined with immune checkpoint inhibitors has demonstrated a gain in pathological complete responses (tpCR) in patients with BC. The aim of our study is to evaluate the safety, feasibility, and efficacy of the addition of dendritic cell vaccines (DCV) to NAC in HER2-negative BC patients.

          Methods:

          Thirty-nine patients with early BC received DCV together with NAC conforming the vaccinated group (VG) and compared with 44 patients as the control group (CG). All patients received anthracyclines and taxanes-based NAC (ddECx4→Dx4) followed by surgery ± radiotherapy ± hormonotherapy.

          Results:

          The tpCR rate was 28.9% in the VG and 9.09% in the CG ( p = 0.03). Pathological CR in the triple negative (TN) BC were 50.0% versus 30.7% ( p = 0.25), 16.6% versus 0% in luminal B ( p = 0.15), and none among luminal A patients in VG versus CG, respectively. Impact of DCV was significantly higher in the programmed cell death ligand 1 (PD-L1) negative population ( p < 0.001). PD-L1 expression was increased in patients with residual disease in the VG as compared with the CG ( p < 0.01). No grade ⩾3 vaccine-related adverse events occurred. With a median follow-up of 8 years, no changes were seen in event-free survival or overall survival. Phenotypic changes post DCV in peripheral blood were observed in myeloid-derived suppressor cells (MDSC), NK, and T cells. Increase in blood cell proliferation and interferon (IFN)-γ production was detected in 69% and 74% in the VG, respectively. Humoral response was also found. Clonality changes in TCR-β repertoire were detected in 67% of the patients with a drop in diversity index after treatment.

          Conclusion:

          The combination of DCV plus NAC is safe and increases tpCR, with a significant benefit among PD-L1-negative tumors. DCV modify tumor milieu and perform cellular and humoral responses in peripheral blood with no impact in outcome.

          Trial registration:

          ClinicalTrials.gov number: NCT01431196. EudraCT 2009-017402-36.

          Related collections

          Most cited references46

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

          Cancer statistics, 2020

          Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on population-based cancer occurrence. Incidence data (through 2016) were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data (through 2017) were collected by the National Center for Health Statistics. In 2020, 1,806,590 new cancer cases and 606,520 cancer deaths are projected to occur in the United States. The cancer death rate rose until 1991, then fell continuously through 2017, resulting in an overall decline of 29% that translates into an estimated 2.9 million fewer cancer deaths than would have occurred if peak rates had persisted. This progress is driven by long-term declines in death rates for the 4 leading cancers (lung, colorectal, breast, prostate); however, over the past decade (2008-2017), reductions slowed for female breast and colorectal cancers, and halted for prostate cancer. In contrast, declines accelerated for lung cancer, from 3% annually during 2008 through 2013 to 5% during 2013 through 2017 in men and from 2% to almost 4% in women, spurring the largest ever single-year drop in overall cancer mortality of 2.2% from 2016 to 2017. Yet lung cancer still caused more deaths in 2017 than breast, prostate, colorectal, and brain cancers combined. Recent mortality declines were also dramatic for melanoma of the skin in the wake of US Food and Drug Administration approval of new therapies for metastatic disease, escalating to 7% annually during 2013 through 2017 from 1% during 2006 through 2010 in men and women aged 50 to 64 years and from 2% to 3% in those aged 20 to 49 years; annual declines of 5% to 6% in individuals aged 65 years and older are particularly striking because rates in this age group were increasing prior to 2013. It is also notable that long-term rapid increases in liver cancer mortality have attenuated in women and stabilized in men. In summary, slowing momentum for some cancers amenable to early detection is juxtaposed with notable gains for other common cancers.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            PD-1 blockade induces responses by inhibiting adaptive immune resistance

            Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. 1–5 One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8 T-cells (termed adaptive immune resistance). 6,7 Here we show that pre-existing CD8 T-cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analyzed samples from 46 patients with metastatic melanoma obtained before and during anti-PD1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next generation sequencing for T-cell receptors (TCR). In serially sampled tumours, responding patients showed proliferation of intratumoural CD8+ T-cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8, PD1, and PD-L1 expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression following therapeutic PD-1 blockade requires pre-existing CD8+ T cells that are negatively regulated by PD-1/PD-L1 mediated adaptive immune resistance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Elements of cancer immunity and the cancer–immune set point

              Immunotherapy is proving to be an effective therapeutic approach in a variety of cancers. But despite the clinical success of antibodies against the immune regulators CTLA4 and PD-L1/PD-1, only a subset of people exhibit durable responses, suggesting that a broader view of cancer immunity is
                Bookmark

                Author and article information

                Contributors
                Journal
                Ther Adv Med Oncol
                Ther Adv Med Oncol
                TAM
                sptam
                Therapeutic Advances in Medical Oncology
                SAGE Publications (Sage UK: London, England )
                1758-8340
                1758-8359
                23 December 2021
                2021
                : 13
                : 17588359211064653
                Affiliations
                [1-17588359211064653]Department of Medical Oncology, Clínica Universidad de Navarra, Avda. Pío XII 36, 31008 Pamplona, Spain
                [2-17588359211064653]Breast Cancer Unit, Clínica Universidad de Navarra, Pamplona, Spain
                [3-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
                [4-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
                [5-17588359211064653]Pharmacometrics and Systems Pharmacology, Universidad de Navarra, Pamplona, Spain
                [6-17588359211064653]Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
                [7-17588359211064653]Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
                [8-17588359211064653]Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
                [9-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Medical Oncology, Complejo Hospitalario de Navarra, Pamplona, Spain
                [10-17588359211064653]Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
                [11-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Department of Preventive Medicine and Public Health, Universidad de Navarra, Pamplona, Spain
                [12-17588359211064653]Breast Cancer Unit, Clínica Universidad de Navarra, Pamplona, Spain
                [13-17588359211064653]Breast Cancer Unit, Clínica Universidad de Navarra, Pamplona, Spain
                [14-17588359211064653]Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
                [15-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
                [16-17588359211064653]Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
                [17-17588359211064653]Cell Therapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
                [18-17588359211064653]Clínica Universidad de Navarra, Universidad de Navarra, Complejo Hospitalario de Navarra and IdisNA, Pamplona, Spain
                [19-17588359211064653]IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
                [20-17588359211064653]Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
                [21-17588359211064653]Cell Therapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
                [22-17588359211064653]Clínica Universidad de Navarra, Universidad de Navarra, Complejo Hospitalario de Navarra and IdisNA, Pamplona, Spain
                Author notes
                [*]

                Equal contribution to the paper.

                Author information
                https://orcid.org/0000-0003-0625-686X
                Article
                10.1177_17588359211064653
                10.1177/17588359211064653
                8721381
                34987618
                dc937ebd-1754-4943-8220-c3430c6e0656
                © The Author(s), 2021

                This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License ( https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages ( https://us.sagepub.com/en-us/nam/open-access-at-sage).

                History
                : 12 August 2021
                : 16 November 2021
                Funding
                Funded by: ministerio de ciencia e innovación, FundRef https://doi.org/10.13039/1501100004837;
                Award ID: PI16/01245
                Funded by: Ministerio de Sanidad y Política Social in the section of Advanced Therapies, ;
                Award ID: TRA-005
                Categories
                Original Research
                Custom metadata
                January-December 2021
                ts1

                dendritic cell vaccines,early breast cancer,immunotherapy,neoadjuvant

                Comments

                Comment on this article