SARS-CoV-2 Vaccine Immunogenicity in Patients with Gastrointestinal Cancer Receiving Systemic Anti-Cancer Therapy

David K Lau, Maria Aresu, Timothy Planche, Amina Tran, Retchel Lazaro-Alcausi, Julie Duncan, Shannon Kidd, Susan Cromarty, Ruwaida Begum, Isma Rana, Su Li, Ali Abdulnabi Mohamed, Irene Monahan, David J Clark, Nicholas Eckersley, Henry M Staines, Elisabetta Groppelli, Sanjeev Krishna, Martin Mayora-Neto, Nigel Temperton, Charlotte Fribbens, David Watkins, Naureen Starling, Ian Chau, David Cunningham, Sheela Rao, David K Lau, Maria Aresu, Timothy Planche, Amina Tran, Retchel Lazaro-Alcausi, Julie Duncan, Shannon Kidd, Susan Cromarty, Ruwaida Begum, Isma Rana, Su Li, Ali Abdulnabi Mohamed, Irene Monahan, David J Clark, Nicholas Eckersley, Henry M Staines, Elisabetta Groppelli, Sanjeev Krishna, Martin Mayora-Neto, Nigel Temperton, Charlotte Fribbens, David Watkins, Naureen Starling, Ian Chau, David Cunningham, Sheela Rao

Abstract

Introduction: Patients with gastrointestinal (GI) cancers have an increased risk of serious complications and death from SARS-CoV-2 infection. The immunogenicity of vaccines in patients with GI cancers receiving anti-cancer therapies is unclear. We conducted a prospective study to evaluate the prevalence of neutralizing antibodies in a cohort of GI cancer patients receiving chemotherapy following SARS-CoV-2 vaccination.

Materials and methods: Between September 2020 and April 2021, patients with cancer undergoing chemotherapy were enrolled. At baseline (day 0), days 28, 56, and 84, we assessed serum antibodies to SARS-CoV-2 spike (anti-S) and anti-nucleocapsid (anti-NP) and concomitantly assessed virus neutralization using a pseudovirus neutralization assay. Patients received either the Pfizer/BioNTech BNT162b2, or the Oxford/AstraZeneca ChAdOx1 vaccine.

Results: All 152 patients enrolled had a prior diagnosis of cancer; colorectal (n = 80, 52.6%), oesophagogastric (n = 38, 25.0%), and hepato pancreatic biliary (n = 22, 12.5%). Nearly all were receiving systemic anti-cancer therapy (99.3%). Of the 51 patients who did not receive a vaccination prior to, or during the study, 5 patients had detectable anti-NP antibodies. Ninety-nine patients received at least one dose of vaccine prior to, or during the study. Within 19 days following the first dose of vaccine, 30.0% had anti-S detected in serum which increased to 70.2% at days 20-39. In the 19 days following a second dose, anti-S positivity was 84.2% (32/38). However, pseudovirus neutralization titers (pVNT80) decreased from days 20 to 39.

Conclusion: Despite the immunosuppressive effects of chemotherapy, 2 doses of SARS-CoV-2 vaccines are able to elicit a protective immune response in patients' ongoing treatment for gastrointestinal cancers. Decreases in pseudoviral neutralization were observed after 20-39 days, re-affirming the current recommendation for vaccine booster doses.

Clinical trial registration number: NCT04427280.

Keywords: COVID-19; SARS-CoV-2; anti-spike; chemotherapy; gastrointestinal cancer; immunity; pseudovirus; vaccines.

© The Author(s) 2022. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
CONSORT diagram of the CARDS study. *Patients RM5287107, RF5287069 withdrew before blood samples were taken. Abbreviations: D0, baseline; D28, day 28; D56, day 56; D84, day 84.
Figure 2.
Figure 2.
Longitudinal analysis of SARS-CoV-2 vaccine immunogenicity relative to the 1st vaccine dose. Box plot of Anti-S index values (A) and pseudovirus neutralisation (titre 1/40) in all vaccinated patients (B) at time periods relative to the date of 1st vaccine dose. Each data point represents a serum sample. Solid horizontal lines denote the median and boxes represent the interquartile range. Samples from patients with positive nucleocapsid results are marked in red and negative in blue. Positivity thresholds are denoted by dotted lines (anti-S > 1.0, pVNT(1/40) > 0.5). Abbreviation: anti-S, anti-spike antibody.
Figure 3.
Figure 3.
Longitudinal analysis of SARS-CoV-2 vaccine immunogenicity following 2nd vaccine dose. Box plot of anti-S index values (A) and pseudovirus neutralisation (titre 1/40) in all vaccinated patients (B) at time periods relative to the date of 2nd SARS-CoV-2 vaccine dose. Each data point represents a serum sample. Solid lines denote the median and boxes represent the interquartile range. Samples from patients with positive nucleocapsid results are marked in red and negative in blue. Prespecified positivity thresholds are denoted by dotted lines (anti-S > 1.0, anti-S > 1.0, pVNT(1/40) > 0.5).
Figure 4.
Figure 4.
Box plots of pVNT80 neutralization titers after the 2nd dose of SARS-CoV-2 vaccine. A logarithmic scale was used for neutralization titre. Each data point represents a serum sample. Solid lines denote the median and boxes represent the interquartile range. Samples from patients with positive nucleocapsid results are marked in red and negative in blue.

References

    1. WHO Coronavirus (COVID-19) Dashboard. Accessed March 3, 2022.
    1. Liang W, Guan W, Chen R, et al. . Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21(3):335-337. 10.1016/S1470-2045(20)30096-6.
    1. Yu J, Ouyang W, Chua MLK, Xie C.. SARS-CoV-2 Transmission in patients with cancer at a tertiary care hospital in Wuhan, China. JAMA Oncol. 2020;6(7):1108-1110. 10.1001/jamaoncol.2020.0980.
    1. Voysey M, Clemens SAC, Madhi SA, et al. . Oxford COVID Vaccine Trial Group. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet (London, England). 2021;397(10269):99-111. 10.1016/S0140-6736(20)32661-1.
    1. Polack FP, Thomas SJ, Kitchin N, et al. . C4591001 Clinical Trial Group. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383(27):2603-2615. 10.1056/nejmoa2034577.
    1. Department of Health and Social Care. Prioritising the first COVID-19 vaccine dose: JCVI statement . Accessed March 3, 2022.
    1. Sung H, Ferlay J, Siegel RL, et al. . Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality Worldwide for 36 cancers in 185 Countries. CA. 2021;71(3):209-249. 10.3322/caac.21660.
    1. Addeo A, Shah PK, Bordry N, et al. . Immunogenicity of SARS-CoV-2 messenger RNA vaccines in patients with cancer. Cancer Cell. 2021;39(8):1091-1098.e2. 10.1016/j.ccell.2021.06.009.
    1. Goshen-Lago T, Waldhorn I, Holland R, et al. . Serologic status and toxic effects of the SARS-CoV-2 BNT162b2 vaccine in patients undergoing treatment for cancer. JAMA Oncol. 2021;7(10):1507-1513. 10.1001/jamaoncol.2021.2675.
    1. Massarweh A, Eliakim-Raz N, Stemmer A, et al. . Evaluation of seropositivity following BNT162b2 messenger RNA vaccination for SARS-CoV-2 in patients undergoing treatment for cancer. JAMA Oncol. 2021;7(8):1133-1140. 10.1001/jamaoncol.2021.2155.
    1. Di Genova C, Sampson A, Scott S, et al. . Production, titration, neutralisation, storage and lyophilisation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) lentiviral pseudotypes. Bio-protocol. 2021;11(21):e4236. 10.21769/BioProtoc.4236.
    1. Hyseni I, Molesti E, Benincasa L, et al. . Characterisation of SARS-CoV-2 lentiviral pseudotypes and correlation between pseudotype-based neutralisation assays and live virus-based micro neutralisation assays. Viruses. 2020;12(9):1011. 10.3390/v12091011.
    1. Thompson CP, Grayson NE, Paton RS, et al. . Detection of neutralising antibodies to SARS-CoV-2 to determine population exposure in Scottish blood donors between March and May 2020. Euro Surveillance: Bull Eur sur les Maladies Transmiss = Eur Commun Dis Bull. 2020;25(42):2000685. 10.2807/1560-7917.es.2020.25.42.2000685.
    1. Hall V, Foulkes S, Insalata F, et al. . Protection against SARS-CoV-2 after Covid-19 vaccination and previous infection. N Engl J Med. 2022;386(13):1207-1220. 10.1056/NEJMoa2118691.
    1. Wei J, Pouwels KB, Stoesser N, et al. . Antibody responses and correlates of protection in the general population after two doses of the ChAdOx1 or BNT162b2 vaccines. Nat Med. 2022;28(5):1072-1082. 10.1038/s41591-022-01721-6.
    1. Monin L, Laing AG, Muñoz-Ruiz M, et al. . Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study. Lancet Oncol. 2021;22(6):765-778. 10.1016/S1470-2045(21)00213-8.
    1. Tran S, Truong TH, Narendran A.. Evaluation of COVID-19 vaccine response in patients with cancer: an interim analysis. Eur J Cancer (Oxford, England: 1990). 2021;159:259-274. 10.1016/j.ejca.2021.10.013.
    1. Thakkar A, Gonzalez-Lugo JD, Goradia N, et al. . Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell. 2021;39(8):1081-1090.e2. 10.1016/j.ccell.2021.06.002.
    1. Naranbhai V, Pernat CA, Gavralidis A, et al. . Immunogenicity and reactogenicity of SARS-CoV-2 vaccines in patients with cancer: the CANVAX Cohort Study. J Clin Oncol. 2022;40(1):12-23. 10.1200/jco.21.01891.
    1. Greenberger LM, Saltzman LA, Senefeld JW, et al. . Antibody response to SARS-CoV-2 vaccines in patients with hematologic malignancies. Cancer Cell. 2021;39(8):1031-1033. 10.1016/j.ccell.2021.07.012.
    1. Bird S, Panopoulou A, Shea RL, et al. . Response to first vaccination against SARS-CoV-2 in patients with multiple myeloma. Lancet Haematol. 2021;8(6):e389-e392. 10.1016/S2352-3026(21)00110-1.
    1. Agha M, Blake M, Chilleo C, Wells A, Haidar G.. Suboptimal response to COVID-19 mRNA vaccines in hematologic malignancies patients. medRxiv: The Preprint Server Health Sci. 2021: 2021.04.06.21254949. 10.1101/2021.04.06.21254949.
    1. Herishanu Y, Avivi I, Aharon A, et al. . Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood. 2021;137(23):3165-3173. 10.1182/blood.2021011568.
    1. Chang A, Akhtar A, Linderman SL, et al. . Humoral responses against SARS-CoV-2 and variants of concern after mRNA vaccines in patients with non-hodgkin lymphoma and chronic lymphocytic leukemia. J Clin Oncol. 2022;40(26):3020-3031. 10.1200/jco.22.00088.
    1. Savage HR, Santos VS, Edwards T, et al. . Prevalence of neutralising antibodies against SARS-CoV-2 in acute infection and convalescence: a systematic review and meta-analysis. PLoS NeglTrop Dis. 2021;15(7):e0009551. 10.1371/journal.pntd.0009551.
    1. Infantino M, Pieri M, Nuccetelli M, et al. . The WHO International Standard for COVID-19 serological tests: towards harmonization of anti-spike assays. Int Immunopharmacol. 2021;100:108095-108095. 10.1016/j.intimp.2021.108095.
    1. Knezevic I, Mattiuzzo G, Page M, et al. . WHO International Standard for evaluation of the antibody response to COVID-19 vaccines: call for urgent action by the scientific community. Lancet Microbe. 2022;3(3):e235-e240. 10.1016/S2666-5247(21)00266-4.
    1. Barrière J, Chamorey E, Adjtoutah Z, et al. . Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann Oncol. 2021;32(8):1053-1055. 10.1016/j.annonc.2021.04.019.
    1. Garcia-Beltran WF, Lam EC, St Denis K, et al. . Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell. 2021;184(9):2372-2383.e9. 10.1016/j.cell.2021.03.013.
    1. Dejnirattisai W, Huo J, Zhou D, et al. ; OPTIC Consortium. SARS-CoV-2 Omicron-B.1.1.529 leads to widespread escape from neutralizing antibody responses. Cell. 2022;185(3):467-484.e15. 10.1016/j.cell.2021.12.046.
    1. Garcia-Beltran WF, St Denis KJ, Hoelzemer A, et al. . mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell. 2022;185(3):457-466.e4. 10.1016/j.cell.2021.12.033.
    1. Earle KA, Ambrosino DM, Fiore-Gartland A, et al. . Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021;39(32):4423-4428. 10.1016/j.vaccine.2021.05.063.

Source: PubMed

3
Abonnieren