but the protective role of these effector mechanisms in
tumor-bearing patients is not clearly established.
Evasion of Immune Responses by Tumors
Immune responses often fail to check tumor growth
because cancers evade immune recognition or resist
immune effector mechanisms. The immune system
faces daunting challenges in combating malignant
tumors, because immune responses must kill all the
tumor cells in order to be effective, and tumors can grow
rapidly. Often, the growth of the tumor simply outstrips
immune defenses. Not surprisingly, tumor cells that
destruction by the immune system (Fig. 10.4):
• Some tumors stop expressing class I MHC molecules
or molecules involved in antigen processing or MHC
assembly, so they cannot display antigens to CD8+ T
evasion than loss of tumor neoantigens because any
tumor may express many immunogenic antigens, all
Fig. 10.3 Immune response against tumors. Tumor antigens are picked up by host dendritic cells and
CHAPTER 10 Immunology of Tumors and Transplantation 201
Production of immunosuppressive
proteins or expression of inhibitory
Mutations in MHC genes or genes
needed for MHC assembly or antigen
Failure to produce tumor antigen
Fig. 10.4 How tumors evade immune responses. Antitumor immunity develops when T cells recognize
MDSC, myeloid derived suppressor cell.
202 CHAPTER 10 Immunology of Tumors and Transplantation
of which would have to be mutated or lost, whereas
mutation in any component of antigen presentation
will lead to failure to present all antigens.
• Tumors engage pathways that inhibit T cell activation. For example, many tumors express PD-L1, a
ligand for the T cell inhibitory receptor programmed
cell death protein 1 (PD-1). Furthermore, tumors,
being persistent, cause repeated stimulation of T
cells specific for tumor antigens. The result is that
the T cells develop an exhausted state, in which they
express high levels of PD-1, cytotoxic T lymphocyte–
associated antigen 4 (CTLA-4), and other inhibitory
molecules, and become unresponsive to antigen.
• Factors in the tumor microenvironment may impair
that capture tumor antigens often express only low
levels of B7 costimulators, resulting in preferential
engagement of the inhibitory receptor CTLA-4 on
naive T cells in the draining lymph nodes, rather
than the stimulatory receptor CD28 (see Chapter 9).
Some tumors may induce regulatory T cells,
which also suppress antitumor immune responses.
Myeloid-derived suppressor cells, which are developmentally related to neutrophils and monocytes but
have mainly antiinflammatory functions, are abundant in tumors, and are believed to contribute to
• Some tumors may secrete immunosuppressive cytokines, such as transforming growth factor ß.
The main strategies for cancer immunotherapy currently
protocols for disseminated cancers, which cannot be cured
surgically, relied on chemotherapy and irradiation, both of
tumors without injuring the patient. Only recently has
the promise of cancer immunotherapy been realized in
patients. The history of cancer immunotherapy illustrates
how the initial, often empirical, approaches have been
largely supplanted by rational strategies based on our
improved understanding of immune responses (Fig. 10.5).
Passive Immunotherapy With Monoclonal
A strategy for tumor immunotherapy which has been in
practice for a limited number of tumors for decades relies
antibodies bind to antigens on the surface of the tumors
(not the neoantigens produced inside cells) and activate
1863 1898 1957 1976 1983 1985 1991 2002 2009 2010 2011 2014
Haanen JB, Punt CJ: Cancer immunotherapy—revisited, Nature Reviews Drug Discovery 10:591–600, 2011.)
CHAPTER 10 Immunology of Tumors and Transplantation 203
host effector mechanisms, such as phagocytes, NK cells,
or the complement system, that destroy the tumor cells.
For example, an antibody specific for CD20, which is
expressed on B cells, is used to treat B cell tumors, usually
in combination with chemotherapy. Although normal
B cells are also depleted, their function can be replaced
the antibody treatment is stopped. Other monoclonal
antibodies that are used in cancer therapy may work by
blocking growth factor signaling (e.g., anti-Her2/Neu for
against the vascular endothelial growth factor for colon
activating the cells ex vivo so there are more of them
and they are more potent effector cells, and transferring
the cells back into the patient. Many variations of this
approach, called adoptive T cell therapy, have been tried.
• Adoptive therapy with autologous tumor-specific
T cells. T cells specific for tumor antigens can be
detected in the circulation and among tumorinfiltrating lymphocytes of cancer patients. T cells
can be isolated from the blood or tumor biopsies of a
patient, expanded by culture with growth factors, and
injected back into the same patient (see Fig. 10-6A).
Presumably, this expanded T cell population contains
activated tumor-specific CTLs, which migrate into the
tumor and destroy it. This approach, which has been
combined with administration of T cell-stimulating
cytokines such as interleukin-2 (IL-2) and traditional
chemotherapy, has shown inconsistent results among
different patients and tumors. One likely reason is that
the frequency of tumor-specific T cells is too low to be
effective in these lymphocyte populations.
• Chimeric antigen receptor (CAR) expressing T cells.
In a more recent modification of adoptive T cell
Passive immunity by transfer of autologous T cells or monoclonal antibodies
Adoptive T cell therapy with CAR-T cells
expanded in vitro Transfer into
gene encoding tumorspecific antigen receptor
204 CHAPTER 10 Immunology of Tumors and Transplantation
Fig. 10.7 Chimeric antigen receptor. The receptor that is
expressed in T cells consists of an extracellular Ig part that
recognizes a surface antigen on tumor cells and intracellular
signaling domains from the TCR complex and costimulatory
receptors that provide the signals that activate the killing function of the T cells.
antigen receptor (CAR), which recognizes a tumor
antigen and provides potent signals to activate the T
cells (see Fig. 10-6B). The CARs currently in use have
a single chain antibody-like extracellular portion with
both heavy- and light-chain variable domains, which
together form the binding site for a tumor antigen
(Fig. 10-7). The specificity of the endogenous T cell
this antibody-based antigen recognition structure
avoids the limitations of MHC restriction of TCRs
cannot evade CAR-T cells by downregulating MHC
expression. In order to work in T cells, the CARs have
intracellular signaling domains of both TCR complex
proteins, for example the ITAMs of the TCR complex
? protein, and the signaling domains of costimulatory
receptors such as CD28 and CD137. Therefore, upon
antigen binding, these receptors provide both antigen
recognition (via the extracellular immunoglobulin
[Ig] domain) and activating signals (via the introduced cytoplasmic domains). CAR-expressing T cells
are expanded ex vivo and transferred back into the
patient, where they recognize the antigen on the tumor
cells and become activated to kill the cells. CAR-T
cell therapy targeting the B cell protein CD19, and
more recently CD20, has shown remarkable efficacy
in treating and even curing B cell-derived leukemias
and lymphomas that are refractory to other therapies.
CARs with other specificities for different tumors are
in development and clinical trials. The most serious
injected T cells recognize and are activated by the
patients’ tumor cells. These cytokines cause high fever,
hypotension, tissue edema, neurologic derangements,
and multi-organ failure. The severity of the syndrome
by on-target, off-tumor toxicities, if the CAR-T cells
are specific for an antigen present on normal cells as
well as tumors. In the case of CD19- or CD20-specific
CARs, the therapy results in depletion of normal B
feasible for other tissues that are destroyed because of
blood (to which the injected T cells have ready access),
it has so far not been successful in solid tumors because
of difficulties in getting T cells into the tumor sites and
the challenge of selecting optimal tumor antigens to
target without injuring normal tissues.
Blocking inhibitory receptors on T cells or their ligands
led to a novel and remarkably effective new strategy for
CHAPTER 10 Immunology of Tumors and Transplantation 205
tumor immunotherapy. The principle of this strategy is to
boost host immune responses against tumors by blocking
normal inhibitory signals for T cells, thus removing the
brakes (checkpoints) on the immune response (Fig. 10.8).
This has been accomplished with blocking monoclonal
antibodies specific for the T cell inhibitory molecules
CTLA-4 and PD-1, first approved for treating metastatic
melanoma in 2011 and 2014, respectively. Since then, the
use of anti-PD-1 or anti-PD-L1 antibodies has expanded
to many different cancer types. The most remarkable
advanced, widely metastatic tumors, which previously
were almost 100% lethal within months to a few years.
tested in clinical trials. There are several novel features
of immune checkpoint blockade and limitations that still
need to be overcome to enhance their usefulness.
• Although the efficacy of checkpoint blockade therapies
for many advanced tumors is superior to any previous
form of therapy, only a subset of patients (25% to 40%
at most) respond to this treatment. The reasons for this
than engaging these inhibitory receptors. Oncologists
CTL-mediated killing of tumor cells
Induction of anti-tumor immune response in lymph node
No costimulation Costimulation
T cell inhibition No T cell inhibition
206 CHAPTER 10 Immunology of Tumors and Transplantation
and immunologists are currently investigating which
biomarkers will predict responsiveness to different
checkpoint blockade approaches.
• One of the most reliable indictors that a tumor will
respond to checkpoint blockade therapy is if it carries a high number of mutations, which correlates
with high numbers of neoantigens and host T cells
that can respond to those antigens. In fact, tumors
that have deficiencies in mismatch repair enzymes,
which normally correct errors in DNA replication
most likely to respond to checkpoint blockade therapy. Remarkably, anti-PD-1 therapy is now approved
for any recurrent or metastatic tumor with mismatch
repair deficiencies, regardless of the cell of origin or
histologic type of tumor. This is a paradigm shift in
how cancer treatments are chosen.
• The combined use of different checkpoint inhibitors,
or one inhibitor with other modes of therapy, will
the combined use of anti-CTLA-4 and anti-PD-1 to
the mechanisms by which CTLA-4 and PD-1 inhibit
T cell activation are different (see Fig. 10.8). There
are numerous ongoing or planned clinical trials using
combinations of checkpoint blockade together with
• The most common toxicities associated with checkpoint blockade are autoimmune damage to organs.
This is predictable, because the physiologic function
of organs may be affected, including colon, lungs,
endocrine organs, heart, and skin, each requiring
different clinical interventions, sometimes including
cessation of the life-saving tumor immunotherapy.
Stimulation of Host Antitumor Immune Responses
by Vaccination With Tumor Antigens
One way of stimulating active immunity against tumors
is to vaccinate patients with their own tumor cells or
prevent infections, tumor vaccines are meant to be
therapeutic, in that they stimulate immune responses to
use these antigens to vaccinate individuals against their
own tumors. Most tumor vaccines tried to date have
No comments:
Post a Comment