Science - USA (2019-01-18)

(Antfer) #1
SCIENCE sciencemag.org

GRAPHIC: N. DESAI/


SCIENCE


after BMT. They propose that passive trans-
fer of antibodies that are matched to the la-
tent CMV strain in HSCT recipients might
constitute a powerful and easy therapeutic
approach to prevent CMV disease.
It is thought that ab T cells, gd T cells,
natural killer (NK) cells, and B cells all con-
tribute to curbing CMV infection ( 6 ) and
that anti-CMV CD8+ T cells are especially
important in CMV immune surveillance and
prevention of reactivation and dissemina-
tion. Analysis of cadaveric organ donor tis-
sue has revealed potential sites of active T
cell–mediated CMV clearance in the blood,
bone marrow, and spleen and reservoirs of
viral persistence in the lung coexisting with
active antiviral T cells ( 7 ). Indeed, transfer of
anti-CMV CD8+ T cells from donors is one
of the approaches investigated to treat CMV
reactivation in immunosuppressed patients
( 8 ). Martins et al. found that in mice, T cell
immunosuppression or depletion alone is
insufficient to enable CMV reactivation after
BMT, and only when host B cells and plasma
cells are eliminated [by graft-versus-host
disease (GVHD), a process by which donor
T cells can attack host cells, or by genetic
deficiency] in addition to T cell and NK cell
depletion does infection emerge (see the fig-
ure). Moreover, they demonstrate that pro-
tection from CMV reactivation conferred
by antibodies is much more efficient if it is
strain specific. Additionally, protection by
antibodies that prevent cell-to-cell dissemi-
nation is more effective than by antibod-
ies that prevent cell entry. Whether these
discoveries in mice—by using a different
conditioning regimen from that used in the
clinic and, by necessity, mouse rather than
human CMV (which are quite different)—
are relevant to the clinical situation merits
careful examination.
Recipients of allogeneic HSCT can be
treated with a variety of regimens to pre-
vent the host immune system from rejecting
the allogeneic stem cells, with a heavy focus
on T cell immunosuppression. These con-
ditioning regimens may include total-body
irradiation at high or low doses, cytoreduc-
ing chemotherapy drugs, and antithymocyte
globulin (ATG) or anti-CD52 (alemtuzumab)
to eliminate host T cells. Given the findings
of Martins et al., it is compelling to con-
sider how these clinical treatments may af-
fect antibody-producing B cells and plasma
cells because CMV reactivation in the clinic,
unlike in mice, can occur in the absence of
GVHD. Plasma cells are thought to be some-
what radioresistant, but ATG is a mixture
of polyclonal antibodies, some of which can
target B cells and plasma cells ( 9 ), and CD52
is expressed on both T and B cells. HSCT
recipients have been reported to experience
prolonged B cell depletion ( 10 ) and might

thus develop a reduction in CMV-specific
antibody titers after HSCT. Prospective fol-
lowing of CMV-specific antibody titers may
be of interest in the clinic to see whether this
is indeed the case.
Prevention or treatment of CMV with im-
munoglobulins has usually been ineffective
in HSCT recipients ( 11 ). The authors argue
that this may be because these reagents,
which come from sera pooled from many
donors, may not contain sufficient concen-
trations of antibodies specific to the CMV
strain infecting a given recipient. That hu-
mans can experience successive infections
with different CMV strains ( 12 ) supports
the possibility that the protective effect of
CMV antibodies is strain specific or at least

that antibodies generated after a given CMV
infection are not broadly cross-protective
against other strains. This is consistent with
the observation of Martins et al. that in their
mouse model, protection against CMV reac-
tivation is only conferred by strain-specific
serum and not when this serum is diluted
with sera containing antibodies reactive
to other strains. If confirmed in the clinic,
generation of a collection of sera specific for
each CMV genotype may be useful for pas-
sive antibody therapy in the future.
An additional point to consider is how
antibodies may work to prevent reactiva-

tion of CMV that is inside infected cells. The
authors show that neutralizing antibodies
that prevent cell infection are not as pro-
tective when injected in vivo as those that
prevent cell-to-cell dissemination in vitro,
which may provide insights for vaccine de-
sign. Much effort has been devoted to de-
veloping vaccines against human CMV ( 13 ).
The study of Martins et al. support giving
further consideration to the role of humoral
immunity in the prevention of reactivation
and dissemination of CMV after clinical
HSCT, the therapeutic potential of strain-
specific serum transfer, and the rational
vaccine design either for CMV-seronegative
recipients or to boost or broaden responses
of seropositive recipients before HSCT. j

REFERENCES


  1. H. Kaminski, J. A. Fishman, Am. J. Transplant. 16 , 2254
    (2016).

  2. D. Furman et al., Sci. Transl. Med. 7 , eaaa2293 (2015).

  3. D. Lilleri, G. Gerna, Immunotherapy 8 , 1135 (2016).

  4. E. Maffini et al., Expert Rev. Hematol. 9 , 585 (2016).

  5. J. P. Martins et al., Science 363 , 288 (2019).

  6. A. Huygens et al., Front. Immunol. 5 , 552 (2014).

  7. C. L. Gordon et al., J. Exp. Med. 214 , 651 (2017).

  8. M. Cobbold et al., J. Exp. Med. 202 , 379 (2005).

  9. M. S. Zand et al., Transplantation 79 , 1507 (2005).

  10. E. Corre et al., Haematologica 95 , 1025 (2010).

  11. P. Raanani et al., J. Clin. Oncol. 27 , 770 (2009).

  12. C. Smith et al., J. Virol. 90 , 7497 (2016).

  13. P. R. Krause et al., Va c c i n e 32 , 4 (2013).


10.1126/science.aav9867

Host infected
fbroblast

CMV antibodies

CMV
dissemination

Host NK cell

Prevention of
CMV disease

Host anti-CMV
CD8+ T cell

Prevention of CMV dissemination

Prevention of CMV dissemination

Decreased CMV
antibodies

GVHD

Prevention of CMV dissemination

Host anti-CMV plasma cell

Host anti-CMV plasma cell

Prevention of CMV dissemination

Host anti-CMV B cell

Host anti-CMV
B cell

Donor alloreactive
CD8+ T cell CMV dissemination CMV disease

18 JANUARY 2019 • VOL 363 ISSUE 6424 233

B cells help prevent CMV reactivation
T cells, NK cells, and B cell–derived antibodies contribute to the prevention of mouse CMV dissemination
after BMT. Killing of host B cells and plasma cells by alloreactive donor T cells (GVHD) in combination with
therapeutic deletion of T cells and NK cells resulted in CMV infection of latently infected hosts.

Published by AAAS

on January 17, 2019^

http://science.sciencemag.org/

Downloaded from
Free download pdf