Seroconversion rates following COVID-19 vaccination among patients with cancer

Astha Thakkar, Jesus D Gonzalez-Lugo, Niyati Goradia, Radhika Gali, Lauren C Shapiro, Kith Pradhan, Shafia Rahman, So Yeon Kim, Brian Ko, R Alejandro Sica, Noah Kornblum, Lizamarie Bachier-Rodriguez, Margaret McCort, Sanjay Goel, Roman Perez-Soler, Stuart Packer, Joseph Sparano, Benjamin Gartrell, Della Makower, Yitz D Goldstein, Lucia Wolgast, Amit Verma, Balazs Halmos, Astha Thakkar, Jesus D Gonzalez-Lugo, Niyati Goradia, Radhika Gali, Lauren C Shapiro, Kith Pradhan, Shafia Rahman, So Yeon Kim, Brian Ko, R Alejandro Sica, Noah Kornblum, Lizamarie Bachier-Rodriguez, Margaret McCort, Sanjay Goel, Roman Perez-Soler, Stuart Packer, Joseph Sparano, Benjamin Gartrell, Della Makower, Yitz D Goldstein, Lucia Wolgast, Amit Verma, Balazs Halmos

Abstract

As COVID-19 adversely affects patients with cancer, prophylactic strategies are critically needed. Using a validated antibody assay against SARS-CoV-2 spike protein, we determined a high seroconversion rate (94%) in 200 patients with cancer in New York City that had received full dosing with one of the FDA-approved COVID-19 vaccines. On comparison with solid tumors (98%), a significantly lower rate of seroconversion was observed in patients with hematologic malignancies (85%), particularly recipients following highly immunosuppressive therapies such as anti-CD20 therapies (70%) and stem cell transplantation (73%). Patients receiving immune checkpoint inhibitor therapy (97%) or hormonal therapies (100%) demonstrated high seroconversion post vaccination. Patients with prior COVID-19 infection demonstrated higher anti-spike IgG titers post vaccination. Relatively lower IgG titers were observed following vaccination with the adenoviral than with mRNA-based vaccines. These data demonstrate generally high immunogenicity of COVID-19 vaccination in oncology patients and identify immunosuppressed cohorts that need novel vaccination or passive immunization strategies.

Keywords: COVID-19; cancer; hematologi malignancies; vaccine.

Conflict of interest statement

Declaration of interests A.V. has received research funding from GlaxoSmithKline, BMS, Janssen, Incyte, MedPacto, Celgene, Novartis, Curis, Prelude, and Eli Lilly and Company, has received compensation as a scientific advisor to Novartis, Stelexis Therapeutics, Acceleron Pharma, and Celgene, and has equity ownership in Stelexis Therapeutics. All other authors declare no competing interests.

Copyright © 2021 Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
CONSORT diagram showing the patient cohort Two hundred and thirteen patients consented to study participation and 29 were enrolled via retrospective chart review. Ultimately based on study criteria, 233 patients were evaluable for vaccine safety analysis and 200 patients were evaluable for vaccine efficacy analysis. One hundred and eighty-five of the 200 patients evaluated for vaccine efficacy analysis were then further assessed as a vaccinated cohort for antibody titer comparisons.
Figure 2
Figure 2
Association of anti-SARS-CoV-2 spike IgG with vaccine types and cancer types (A) Patients with hematologic malignancies had lowest titers when compared with those with solid tumors and non-cancer patient controls. No difference was seen between patients with solid tumors and controls. (B) Anti-spike protein IgG antibody titers (AU/mL) were significantly higher in patients who received mRNA vaccines than in those who received adenoviral vaccine. Box plots here and in subsequent figures show median (horizontal bar), the 75th and 25th quartiles, and error bars depicting the largest and smallest values (up to 1.5 times the interquartile range). Differences assessed by Kruskal-Wallis test.
Figure 3
Figure 3
Association of anti-SARS-CoV-2 spike IgG with therapy (A–C) Anti-spike protein IgG antibody titers (AU/mL) after full vaccination did not significantly differ in patients receiving active therapy (A), chemotherapy (B), or radiation therapy (C) when compared with respective counterparts. (D)Patients that had received surgery versus no surgery had no significant difference in titer levels (p = 0.08). Box plots are shown with differences assessed by Kruskal-Wallis test.
Figure 4
Figure 4
Association of anti-SARS-CoV-2 spike IgG with immunosuppressive therapies (A and B) Anti-spike protein IgG antibody titers (AU/mL) after full vaccination did not significantly differ in patients having received stem cell transplantation (SCT) (A) or anti-CD38 antibody therapy (B) when compared with respective counterparts. (C and D) Patients receiving anti-CD20 antibody treatments (C) or CAR-T cell therapy (D) had a significantly lower titer after vaccination when compared with respective counterparts. Box plots are shown with differences assessed by Kruskal-Wallis test.
Figure 5
Figure 5
Association of anti-SARS-CoV-2 spike IgG with type of therapy and prior COVID-19 history (A) Anti-spike protein IgG antibody titers (AU/mL) after full vaccination did not significantly differ in patients receiving immune checkpoint inhibitor (ICI) therapy when compared with those who did not. (B) Patients receiving hormonal therapy had a significantly higher titer after vaccination. (C) Patients receiving CDK4/6 inhibitor therapy had a significantly lower titer after vaccination. (D) Patients with prior COVID-19 infection had significantly higher IgG titers. Box plots are shown with differences assessed by Kruskal-Wallis test.

References

    1. Baden L.R., El Sahly H.M., Essink B., Kotloff K., Frey S., Novak R., Diemert D., Spector S.A., Rouphael N., Creech C.B., et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2020;384:403–416.
    1. Bakouny Z., Hawley J.E., Choueiri T.K., Peters S., Rini B.I., Warner J.L., Painter C.A. COVID-19 and cancer: current challenges and perspectives. Cancer Cell. 2020;38:629–646.
    1. Bange E.M., Han N.A., Wileyto P., Kim J.Y., Gouma S., Robinson J., Greenplate A.R., Hwee M.A., Porterfield F., Owoyemi O., et al. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat. Med. 2021 doi: 10.1038/s41591-021-01386-7.
    1. Barrière J., Chamorey E., Adjtoutah Z., Castelnau O., Mahamat A., Marco S., Petit E., Leysalle A., Raimondi V., Carles M. Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann. Oncol. 2021 doi: 10.1016/j.annonc.2021.04.019.
    1. Bird S., Panopoulou A., Shea R.L., Tsui M., Saso R., Sud A., West S., Smith K., Barwood J., Kaczmarek E., et al. Response to first vaccination against SARS-CoV-2 in patients with multiple myeloma. Lancet Haematol. 2021;8:e389–e392.
    1. Bradley B.T., Bryan A., Fink S.L., Goecker E.A., Roychoudhury P., Huang M.-L., Zhu H., Chaudhary A., Madarampalli B., Lu J.Y.C., et al. Anti-SARS-CoV-2 antibody levels are concordant across multiple platforms but are not fully predictive of sterilizing immunity. medRxiv. 2021 doi: 10.1101/2021.04.26.21256118.
    1. Chen-Kiang S. Cell-cycle control of plasma cell differentiation and tumorigenesis. Immunol. Rev. 2003;194:39–47.
    1. Cingöz O., Goff S.P. Cyclin-dependent kinase activity is required for type I interferon production. Proc. Natl. Acad. Sci. 2018;115:E2950–E2959.
    1. Desai A., Gainor J.F., Hegde A., Schram A.M., Curigliano G., Pal S., Liu S.V., Halmos B., Groisberg R., Grande E., et al. COVID-19 vaccine guidance for patients with cancer participating in oncology clinical trials. Nat. Rev. Clin. Oncol. 2021;18:313–319.
    1. Dhand A., Lobo S.A., Wolfe K., Feola N., Nabors C. Bamlanivimab for treatment of COVID-19 in solid organ transplant recipients: early single-center experience. Clin. Transplant. 2021;35:e14245.
    1. Friese C.R., Choueiri T.K., Duma N., Farmakiotis D., Grivas P., Rini B.I., Shah D.P., Thompson M.A., Pergam S.A., Mishra S., Warner J.L. Care without a compass: including patients with cancer in COVID-19 studies. Cancer Cell. 2021 doi: 10.1016/j.ccell.2021.04.006.
    1. García-Suárez J., De La Cruz J., Cedillo Á., Llamas P., Duarte R., Jiménez-Yuste V., Hernández-Rivas J.Á., Gil-Manso R., Kwon M., Sánchez-Godoy P., et al. Impact of hematologic malignancy and type of cancer therapy on COVID-19 severity and mortality: lessons from a large population-based registry study. J. Hematol. Oncol. 2020;13:133.
    1. Grivas P., Khaki A.R., Wise-Draper T.M., French B., Hennessy C., Hsu C.Y., Shyr Y., Li X., Choueiri T.K., Painter C.A., et al. Association of clinical factors and recent anticancer therapy with COVID-19 severity among patients with cancer: a report from the COVID-19 and Cancer Consortium. Ann. Oncol. 2021;32:787–800.
    1. Herishanu Y., Avivi I., Aharon A., Shefer G., Levi S., Bronstein Y., Morales Moshiashvili M., Ziv-Baran T., Shorer Y., Scarfo L., et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood. 2021 doi: 10.1182/blood.2021011568.
    1. Hurt A.C., Wheatley A.K. Neutralizing antibody therapeutics for COVID-19. Viruses. 2021;13:628.
    1. Jaffe D., Papadopoulos E.B., Young J.W., O'reilly R.,J., Prockop S., Kernan N.A., Jakubowski A., Boulad F., Perales M.A., Castro-Malaspina H., Small T.N. Immunogenicity of recombinant hepatitis B vaccine (rHBV) in recipients of unrelated or related allogeneic hematopoietic cell (HC) transplants. Blood. 2006;108:2470–2475.
    1. Jee J., Foote M.B., Lumish M., Stonestrom A.J., Wills B., Narendra V., Avutu V., Murciano-Goroff Y.R., Chan J.E., Derkach A., et al. Chemotherapy and COVID-19 outcomes in patients with cancer. J. Clin. Oncol. 2020;38:3538–3546.
    1. Keam B., Kim M.K., Choi Y., Choi S.J., Choe P.G., Lee K.H., Kim T.M., Kim T.Y., Oh D.Y., Kim D.W., et al. Optimal timing of influenza vaccination during 3-week cytotoxic chemotherapy cycles. Cancer. 2017;123:841–848.
    1. Krammer F., Srivastava K., Alshammary H., Amoako A.A., Awawda M.H., Beach K.F., Bermúdez-González M.C., Bielak D.A., Carreño J.M., Chernet R.L., et al. Antibody responses in seropositive persons after a single dose of SARS-CoV-2 mRNA vaccine. N. Engl. J. Med. 2021;384:1372–1374.
    1. Kuderer N.M., Choueiri T.K., Shah D.P., Shyr Y., Rubinstein S.M., Rivera D.R., Shete S., Hsu C.-Y., Desai A., De Lima Lopes G., et al. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. The Lancet. 2020;395:1907–1918.
    1. Laphanuwat P., Jirawatnotai S. Immunomodulatory roles of cell cycle regulators. Front. Cell. Dev. Biol. 2019;7:23.
    1. Lee L.Y., Cazier J.-B., Angelis V., Arnold R., Bisht V., Campton N.A., Chackathayil J., Cheng V.W., Curley H.M., Fittall M.W., et al. COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: a prospective cohort study. Lancet. 2020;395:1919–1926.
    1. Lee L.Y.W., Cazier J.-B., Starkey T., Briggs S.E.W., Arnold R., Bisht V., Booth S., Campton N.A., Cheng V.W.T., Collins G., et al. COVID-19 prevalence and mortality in patients with cancer and the effect of primary tumour subtype and patient demographics: a prospective cohort study. Lancet Oncol. 2020;21:1309–1316.
    1. Marra A., Generali D., Zagami P., Cervoni V., Gandini S., Venturini S., Morganti S., Passerini R., Orecchia R., Curigliano G. Seroconversion in patients with cancer and oncology health care workers infected by SARS-CoV-2. Ann. Oncol. 2020;32:113–119.
    1. Mehta V., Goel S., Kabarriti R., Cole D., Goldfinger M., Acuna-Villaorduna A., Pradhan K., Thota R., Reissman S., Sparano J.A., et al. Case fatality rate of cancer patients with COVID-19 in a New York hospital system. Cancer Discov. 2020;10:935–941.
    1. Monin-Aldama L., Laing A.G., Muñoz-Ruiz M., Mckenzie D.R., Del Molino Del Barrio I., Alaguthurai T., Domingo-Vila C., Hayday T.S., Graham C., Seow J., et al. Interim results of the safety and immune-efficacy of 1 versus 2 doses of COVID-19 vaccine BNT162b2 for cancer patients in the context of the UK vaccine priority guidelines. medRxiv. 2021 doi: 10.1101/2021.03.17.21253131.
    1. Pillay T.S. Gene of the month: the 2019-nCoV/SARS-CoV-2 novel coronavirus spike protein. J. Clin. Pathol. 2020;73:366–369.
    1. Pleyer C., Ali M.A., Cohen J.I., Tian X., Soto S., Ahn I.E., Gaglione E.M., Nierman P., Marti G.E., Hesdorffer C., et al. Effect of Bruton tyrosine kinase inhibitor on efficacy of adjuvanted recombinant hepatitis B and zoster vaccines. Blood. 2021;137:185–189.
    1. Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Pérez Marc G., Moreira E.D., Zerbini C., et al. Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615.
    1. Ribas A., Sengupta R., Locke T., Zaidi S.K., Campbell K.M., Carethers J.M., Jaffee E.M., Wherry E.J., Soria J.C., D'souza G. Priority COVID-19 vaccination for patients with cancer while vaccine supply is limited. Cancer Discov. 2021;11:233–236.
    1. Rizk J.G., Forthal D.N., Kalantar-Zadeh K., Mehra M.R., Lavie C.J., Rizk Y., Pfeiffer J.P., Lewin J.C. Expanded access programs, compassionate drug use, and emergency use authorizations during the COVID-19 pandemic. Drug Discov. Today. 2021;26:593–603.
    1. Robilotti E.V., Babady N.E., Mead P.A., Rolling T., Perez-Johnston R., Bernardes M., Bogler Y., Caldararo M., Figueroa C.J., Glickman M.S., et al. Determinants of COVID-19 disease severity in patients with cancer. Nat. Med. 2020;26:1218–1223.
    1. Rubin L.G., Levin M.J., Ljungman P., Davies E.G., Avery R., Tomblyn M., Bousvaros A., Dhanireddy S., Sung L., Keyserling H., Kang I. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin. Infect. Dis. 2014;58:309–318.
    1. Sadoff J., Gray G., Vandebosch A., Cárdenas V., Shukarev G., Grinsztejn B., Goepfert P.A., Truyers C., Fennema H., Spiessens B., et al. Safety and efficacy of single-dose Ad26.COV2.S vaccine against covid-19. N. Engl. J. Med. 2021 doi: 10.1056/NEJMoa2101544.
    1. Terpos E., Trougakos I.P., Gavriatopoulou M., Papassotiriou I., Sklirou A.D., Ntanasis-Stathopoulos I., Papanagnou E.D., Fotiou D., Kastritis E., Dimopoulos M.A. Low neutralizing antibody responses against SARS-CoV-2 in elderly myeloma patients after the first BNT162b2 vaccine dose. Blood. 2021 doi: 10.1182/blood.2021011904.
    1. Thakkar A., Pradhan K., Jindal S., Cui Z., Rockwell B., Shah A.P., Packer S., Sica R.A., Sparano J., Goldstein D.Y., et al. Patterns of seroconversion for SARS-CoV-2 IgG in patients with malignant disease and association with anticancer therapy. Nat. Cancer. 2021;2:392–399.
    1. Van Der Veldt A.A.M., Oosting S.F., Dingemans A.C., Fehrmann R.S.N., Geurtsvankessel C., Jalving M., Rimmelzwaan G.F., Kvistborg P., Blank C.U., Smit E.F., et al. COVID-19 vaccination: the VOICE for patients with cancer. Nat. Med. 2021;27:568–569.
    1. Waissengrin B., Agbarya A., Safadi E., Padova H., Wolf I. Short-term safety of the BNT162b2 mRNA COVID-19 vaccine in patients with cancer treated with immune checkpoint inhibitors. Lancet Oncol. 2021;22:581–583.
    1. Westblade L.F., Brar G., Pinheiro L.C., Paidoussis D., Rajan M., Martin P., Goyal P., Sepulveda J.L., Zhang L., George G., et al. SARS-CoV-2 viral load predicts mortality in patients with and without cancer who are hospitalized with COVID-19. Cancer Cell. 2020;38:661–671.e2.
    1. Widge A.T., Rouphael N.G., Jackson L.A., Anderson E.J., Roberts P.C., Makhene M., Chappell J.D., Denison M.R., Stevens L.J., Pruijssers A.J., et al. Durability of responses after SARS-CoV-2 mRNA-1273 vaccination. N. Engl. J. Med. 2020;384:80–82.
    1. Yang J., Petitjean S.J.L., Koehler M., Zhang Q., Dumitru A.C., Chen W., Derclaye S., Vincent S.P., Soumillion P., Alsteens D. Molecular interaction and inhibition of SARS-CoV-2 binding to the ACE2 receptor. Nat. Commun. 2020;11:4541.

Source: PubMed

3
Iratkozz fel