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
. .

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
. .

Van Oekelen
O
,
Gleason
CR
,
Agte
S
, et al
.
Highly variable SARS-CoV-2 spike antibody responses to two doses of COVID-19 RNA vaccination in patients with multiple myeloma
.
Cancer Cell
.
2021
;
39
(
8
):
1028
1030
. .

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
. .

Patients with cancer have disproportionately adverse outcomes after COVID-19 infection, experiencing more severe complications from infection and also significant interruptions to delivery of standard clinical care.1  Vaccination strategies against the SARS-CoV-2 virus are highly efficacious in protection against severe complications of disease; however, SARS-CoV-2 vaccine efficacy trials have excluded patients with cancer.25  Recently, numerous important cohort studies have examined mRNA vaccine regimens in patients with blood cancers, specifically asking whether immunogenicity is retained in cancer patients assessed by the evaluation of qualitative or quantitative antibody response to the SARS-CoV-2 spike protein. This work extrapolates from early findings demonstrating that vaccine efficacy is linked to antibody neutralization titer.6  Importantly, while these studies rely on humoral responses to vaccination, there is likely to be a contribution from other arms of the immune system — a question for future research.

In aggregate, there are three main messages from these articles. The first is that as a cohort, patients with hematologic cancer have lower rates of anti-spike IgG seropositivity and lower titers when compared to a solid organ cancer population and non-cancer controls. In these instances, the rate of seropositivity was 85 percent versus 98 percent versus 100 percent, respectively. Although this was not examined in a randomized manner, the highest titers of vaccine responses were seen after mRNA1273 (Moderna), followed by BNT162b2 (BioNTech/Pfizer), followed by a single dose of Ad26.COV2.S (Janssen). This comparison needs to be examined by prospective studies. Interestingly, patients who previously had documented COVID infection seemed to have higher antibody titers, supporting a role for immune priming prior to vaccination.

The second common theme was that there are several specific diseases where spike antibody seroconversion and titers are most impaired. These included non-Hodgkin lymphoma, multiple myeloma, and chronic lymphocytic leukemia (CLL) — all B-cell malignancies with high rates of hypogammaglobulinemia and impaired humoral immune responses. Specifically, antibodies to SARS-CoV-2 spike protein were detected in 40 percent (66/167) of patients with CLL after two doses of mRNA vaccine.7  This result mirrors the findings of another study where just 64 percent (417/650) of patients with CLL demonstrated seropositivity after two doses of mRNA vaccine. In the large cohort observed by Dr. Lee M. Greenberger and colleagues, patients with B-cell non-Hodgkin lymphoma had the highest risk of seronegativity. Finally, in two cohorts of patients with multiple myeloma, spike-protein antibodies were detected after two doses of vaccine in 84 percent (219/260) and 95 percent (175/184) of assessed patients. Conversely, seropositivity was seen in high rates for some conditions, including Hodgkin lymphoma (99%; 64/65), chronic myeloid leukemia (97%; 33/34), acute myeloid leukemia (91%; 31/34), and acute lymphoblastic leukemia (88%; 15/17).

The final common theme is that the choice of cancer treatment can have a profound impact on the rates of seroconversion. Seropositive rates were particularly low in patients treated with monoclonal antibodies that deplete B cells (rituximab 44%; 84/191), Bruton tyrosine kinase inhibitors (BTKi; zanubrutinib 50%, n=4; acalabrutinib 43%, n=56; ibrutinib 49%, n=222), and CD19 chimeric antigen receptor T-cell therapy (14%, n=7). In a CLL cohort, none of the 22 patients that received anti-CD20 therapy in the 12 months prior to vaccination had a detectable serological response. Of those with at least 12 months between anti-CD20 therapy and vaccination, 46 percent (25/55) demonstrated serological response. These rates compare to 100 percent seropositivity in an immunocompetent population. These findings may have substantial implications for patients on maintenance B-cell–depleting therapy and therefore need careful reassessment of the perceived risk versus benefit of these strategies during this current pandemic.

These variable humoral responses to vaccination raise the question as to what more can be done. Avoidance of exposure remains a top priority in these high-risk populations. Regulators in the United States and other countries have recommended an additional booster in some immunocompromised groups per the Centers for Disease Control and Prevention. This recommendation follows data demonstrating improved serological response to third doses of mRNA vaccine in solid organ transplant recipients and renal dialysis patients.7-9  Data such as these are eagerly awaited in other immunocompromised cohorts. Pre-print data in hematological cancer cohorts reported ongoing seronegativity in those who failed to respond after a two-dose mRNA regimen, despite a third mRNA booster.10  Another potential consideration may be heterologous boosting, or the use of a second distinct vaccine stimulus to augment the initial vaccine. Along these lines, a patient actively receiving B-cell–targeting therapy (prior anti-CD20, followed by current BTKi therapy) failed to produce detectable anti-SARS-CoV-2 spike IgG after two doses of mRNA vaccine, but then produced detectable (but low titer) antibodies after boosting with Ad26.COV2.S viral vector vaccine.11 

Figure:

High-risk patients and mRNA COVID vaccine response. Patients with B-cell malignancies and who have had chronic B-cell–depleting therapies have reduced response to mRNA COVID-19 vaccines. Strategies to protect these patients include rigorous public exposure avoidance, early intervention to try and reduce disease severity, minimizing exposure to chronic B-cell depletion, and third dose vaccine strategies.

Figure:

High-risk patients and mRNA COVID vaccine response. Patients with B-cell malignancies and who have had chronic B-cell–depleting therapies have reduced response to mRNA COVID-19 vaccines. Strategies to protect these patients include rigorous public exposure avoidance, early intervention to try and reduce disease severity, minimizing exposure to chronic B-cell depletion, and third dose vaccine strategies.

Close modal

These data provide limited information about the efficacy of adenovirus vaccines in these populations, and further studies are required to help guide vaccine choice in countries with access to both adenovirus and mRNA vaccines. Furthermore, although vaccine titers seem to correlate with protection against severe disease, more follow up is required to determine if this finding holds true for immunocompromised populations. Further studies in immunocompromised and high-risk cohorts, such as those with blood cancers, are urgently needed to guide vaccination and disease minimization strategies and to help identify patients who need rapid escalation of specific COVID-19 treatments.

Dr. Michael Lane and Dr. Steven Lane indicated no relevant conflicts of interest.

1.
Bakouny
Z
,
Hawley
JE
,
Choueiri
TK
, et al
.
COVID-19 and cancer: current challenges and perspectives
.
Cancer Cell
.
2020
;
38
(
5
):
629
646
.
2.
Sadoff
J
,
Gray
G
,
Vandebosch
A
, et al
.
Safety and efficacy of single-dose Ad26.COV2.S vaccine against COVID-19
.
N Engl J Med
.
2021
;
384
(
23
):
2187
2201
.
3.
Voysey
M
,
Costa Clements
SA
,
Madhi
SA
, et al
.
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
.
2021
;
397
(
10269
):
99
11
.
4.
Polack
FP
,
Thomas
SJ
,
Kitchin
N
, et al
.
Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine
.
N Engl J Med
.
2020
;
383
(
27
):
2603
2615
.
5.
Baden
LR
,
El Sahly
HM
,
Essink
B
, et al
.
Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine
.
N Engl J Med
.
2021
;
384
(
5
):
403
416
.
6.
Krammer
F
.
A correlate of protection for SARS-CoV-2 vaccines is urgently needed
.
Nat Med
.
2021
;
27
(
7
):
1147
1148
.
7.
Hall
VG
,
Ferreira
VH
,
Ku
T
, et al
.
Randomized trial of a third dose of mRNA-1273 vaccine in transplant recipients
.
N Engl J Med
.
2021
;
385
(
13
):
1244
1246
.
8.
Benotmane
I
,
Gautier
G
,
Perrin
P
, et al
.
Antibody response after a third dose of the mRNA-1273 SARS-CoV-2 vaccine in kidney transplant recipients with minimal serologic response to 2 doses
.
JAMA
.
2021
;
326
(
11
):
1063
1065
.
9.
Longlune
N
,
Nogier
MB
,
Miedougé
M
, et al
.
High immunogenicity of a messenger RNA-based vaccine against SARS-CoV-2 in chronic dialysis patients
.
Nephrol Dial Transplant
.
2021
;
36
(
9
):
1704
1709
.
10.
Re
D
,
Seitz-Polski
B
,
Carles
M
, et al
.
Humoral and cellular responses after a third dose of BNT162b2 vaccine in patients treated for lymphoid malignancies
.
medRxiv
.
2021
; DOI: .
11.
Hill
JA
,
Ujjani
CS
,
Greninger
AL
, et al
.
Immunogenicity of a heterologous COVID-19 vaccine after failed vaccination in a lymphoma patient
.
Cancer Cell
.
2021
;
39
(
8
):
1037
1038
.