Accepted Manuscript Considerations for successful cancer immunotherapy in aged hosts
Vincent Hurez, Álvaro Padrón, Robert S. Svatek, Tyler J. Curiel PII: DOI: Reference:
S0531-5565(17)30415-1 doi:10.1016/j.exger.2017.10.002 EXG 10167
To appear in:
Experimental Gerontology
Received date: Revised date: Accepted date:
30 May 2017 30 September 2017 3 October 2017
Please cite this article as: Vincent Hurez, Álvaro Padrón, Robert S. Svatek, Tyler J. Curiel , Considerations for successful cancer immunotherapy in aged hosts. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Exg(2017), doi:10.1016/j.exger.2017.10.002
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Considerations for successful cancer immunotherapy in aged hosts Vincent Hurez1, Álvaro Padrón1, Robert S. Svatek2,3, Tyler J. Curiel1,3-5,*
1
Department of Medicine, University of Texas Health San Antonio, TX 78229
2
Department of Urology, University of Texas Health San Antonio, TX 78229
3
T
The UT Health Cancer Center, University of Texas Health San Antonio, TX 78229
4
IP
Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, TX
CR
78229 5
AN
Running title: Aging and cancer immunotherapy
US
The Barshop Institute for Aging and Longevity Studies, University of Texas Health San Antonio TX 78229
Keywords: Aging, immunity, immunotherapy, cancer
M
Abbreviations used: ARID, Age Related Immune Dysfunction; BCG, Bacille Calmette-Guérin; CAR, chimeric
ED
antigen receptor; CDMRP, Congressionally Directed Medical Research Program; CD40L, CD40 ligand; CpG, poly-(cysteine 5’ to guanine); CRISPR, clustered regularly interspaced short palindromic repeats; CTLA-4,
PT
cytotoxic T lymphocyte antigen-4; DC, dendritic cell; DNA, deoxyribonucleic acid; ICAM-1, intracellular
CE
adhesion molecule-1; IL, interleukin; Lag3, lymphocyte activation gene 3; M1, type 1 (macrophage); M2, type 2 (macrophage); MDSC, myeloid derived suppressor cell; MHC, major histocompatibility complex; mTOR,
AC
mammalian target of rapamycin; PD-1, programmed death-1; PD-L1, programmed death ligand-1; Th17, T helper cell 17; TGF-β, transforming growth factor-β; Tim3, T-cell immunoglobulin and mucin-domain containing-3; TNF-α, tumor necrosis factor-α; Treg, regulatory T cell
Financial support: Tyler Curiel (CA170491, CA54174, CDMRP, The Holly Beach Public Library, The Owens Foundation, The Barker Foundation and the Skinner endowment).
1
ACCEPTED MANUSCRIPT *To whom correspondence should be addressed: Tyler Curiel, MD, MPH, Department of Medicine, University of Texas Health San Antonio, STRF MC 8252, 8403 Floyd Curl Drive, San Antonio, TX 78229-3900, USA. Phone: 210-562-4083, E-mail:
[email protected]
Abstract
T
Improvements in understanding cancer immunopathogenesis has now led to unprecedented successes in
IP
immunotherapy to treat numerous cancers. Although aging is the most important risk factor for cancer, most
CR
pre-clinical cancer immunotherapy studies are undertaken in young hosts. This review covers age-related immune changes as they affect cancer immune surveillance, immunopathogenesis and immune therapy
US
responses. Declining T cell function with age can impede efficacy of age-related cancer immunotherapies, but examples of successful approaches to breach this barrier have been reported. It is further recognized now that
AN
immune functions with age do not simply decline, but that they change in potentially detrimental ways. For example, detrimental immune cell populations can become predominant during aging (notably pro-
M
inflammatory cells), the prevalence or function of suppressive cells can increase (notably myeloid derived
ED
suppressor cells), drugs can have age-specific effects on immune cells, and attributes of the aged microenvironment can impede or subvert immunity. Key advances in these and related areas will be reviewed
PT
as they pertain to cancer immunotherapy in the aged, and areas requiring additional study and some
CE
speculations on future research directions will be addressed. We prefer the term Age Related Immune
AC
Dysfunction (ARID) as most encompassing the totality of age-associated immune changes.
2
ACCEPTED MANUSCRIPT 1.0 INTRODUCTION The immune system can identify (and eliminate) cells expressing specific antigens with remarkable sensitivity. Tumors are theoretically highly antigenic tissues owing to their various mutations. Antigenicity, the expression of antigens, nonetheless does necessarily equal clinically useful immunogenicity (the capacity of antigens to provoke useful immunity). Thus, spontaneous rejection of clinically apparent tumors is a rare event. Work over
T
the past 15 years shows how endogenous immunity fails to eradicate clinically through a multiplicity of factors
IP
including immunoediting (discussed below), from tumor-driven inflammation and immune dysfunction, and
CR
because tumor rejection antigens can be self antigens that are protected by autoimmune defense mechanisms [1, 2]. Recent work has advanced thinking towards anti-cancer immunotherapies with high potential for
US
efficacy, which has now clearly been demonstrated clinically. As hosts age, these already-formidable barriers
AN
to treatment success become compounded, and additional barriers can emerge [3-6].
Tumor immune surveillance is part of larger scheme of immunoediting [7], easily remember as involving three
M
E’s [8]. The first “E” is elimination of cancer cells as they initially emerge. The second “E” is equilibrium, in
ED
which malignant cells mutate to evolve immune elimination as the immune system evolves to attack new mutations. Selective pressure during equilibrium provoke tumor antigenic evolution that finally allows immune
PT
escape, the third “E”. In escape, the tumor can evade immune defenses and become clinically manifest, a
CE
process seen in mouse models [9-11] and in humans[12-14].
AC
The original six fundamental cancer hallmarks [15] did not include lack of immune rejection, which was corrected with the revision to eight fundamental hallmarks [16]. Generalized, chronic inflammation [17, 18] is also considered a cancer hallmark [19], as well as stem cell features [20], genomic instability [21], and abnormal vasculature [22].
These cancer hallmarks (particularly immune rejection and chronic inflammation) affect age-specific immunotherapy development. Age effects on immunity extend far beyond simple declines in functions or reductions in cell numbers. We propose the term “Age Related Immune Dysfunction” (ARID) to encompass the 3
ACCEPTED MANUSCRIPT full range of age-related alterations in immunity with advancing age. The following sections address major topics relating to age effects on cancer immunotherapy.
1.1 TUMOR-SPECIFIC T CELLS T cell immunity wans with age owing to various processes affecting T cell numbers, diversity, phenotype and
T
function [23]. As an example, CD45RA+CD62L+CD27+CD28+ naive T cells are generated in thymus throughout
IP
life, but production decreases with age [24], and thus T cell repertoire diversity declines with age as overall
CR
peripheral T cell numbers stay essentially the same [25]. Hematopoietic stem cell generation of T cell precursors [26] also declines. While naïve T cells decrease, there is a concomitant increase in memory T cells
US
with age. Apparently terminally differentiated effector T cells (based on flow cytometry phenotype), including virus-reactive cells, with greatly reduced T cell receptor repertoire diversity and compromised potential to
AN
proliferate specifically increase [27]. T cell signaling, including through the T cell receptor also declines with
M
age [28].
ED
Strategies to reduce age-related defects and improve T cell immunity have been tested. CD4+ T cell functions decline with age, but such loss can be partially overcome using cytokines including tumor necrosis factor
PT
(TNF)-α or interleukin (IL)-6 [29]. T cell priming (the activation of naïve, antigen-specific T cells) is defective in
CE
age, but can be improved with agonist αCD137 antibodies [30]. Tumor-specific immunity can also be improved. For example, tumor control and anti-tumor T cell differentiation through OX40 signals decreases with age [31,
AC
32], but aged mice can be induced to mount protective antitumor immunity in a lymphoma model using an agonist αCD40 antibody [33]. We found that aged mice develop anti-tumor immunity comparable to that in young mice against B16 melanoma, a poorly immunogenic and highly aggressive tumor, by simultaneous reduction of age-increased suppressive myeloid and dysfunctional regulatory T cells [5], which is discussed in detail below.
4
ACCEPTED MANUSCRIPT T cells (not necessarily cancer-specific) can improve host response to anti-cancer immunotherapy [34]. It has been proposed that improving the function of aging T cells could improve cancer chemotherapy responses in the aged [35], although this concept has not been tested to our knowledge.
1.2 REGULATORY T CELLS
T
Regulatory T cells (Tregs) mediate significant tumor immune dysfunction. Thus, inhibiting Treg function or
IP
reducing their numbers is under study as cancer immunotherapy [36-39]. Reports of Treg effects in age-related
CR
immune dysfunction are contradictory. Some studies showing show no changes or lessened Treg contributions [40, 41], whereas other studies find increased age-related Treg function and/or prevalence in humans and
US
mice [42-45]. Treg prevalence in lymphoid organs but not thymus or blood have been reported in aged mice
AN
[5, 46, 47].
Age effects on Treg functions depends on context and the specific function investigated. Some functions could
M
be reduced [48], such as suppressing delayed type hypersensitivity [44], or inhibiting Th17 immunity [46]. Aged
ED
Tregs mediate equal or greater suppression versus young mice in some reports [5, 49]. Treg data in elderly humans is limited, but they could increase in with age in blood [50, 51]. Tregs from young and elderly persons
CE
PT
inhibited T cell proliferation similarly, but IL- 10 (an anti-inflammatory cytokine) was lower in aged Tregs [52].
Treg depletion can improve anti-tumor immunity and immunotherapy outcomes [37, 39], but reported Treg
AC
depletion effects in cancer immunotherapy in aged hosts are conflicting [53, 54]. One mouse study linked impaired tumor rejection to increased Tregs. In this study, αCD25 depleted Tregs and improved anti-cancer immunity [54]. We depleted Tregs in mice bearing B16 melanoma using denileukin diftitox [5]. Denileukin diftitox similarly depleted Tregs in young and aged mice, but it delayed tumor growth and augmented tumorspecific immunity only in young mice. Denileukin diftitox altered interferon-γ and IL-17 producing T cells distinctly in young versus aged mice. CD11b+Gr-1hi myeloid derived suppressor cells (MDSC) were more numerous and suppressive in aged tumor-bearing aged mice, and depleting Tregs increased MDSC numbers even further. 5
ACCEPTED MANUSCRIPT We added anti-Gr-1 antibody to deplete MDSC along with Treg depletion, and restored anti-tumor immunity in the aged mice that slowed tumor growth comparable to Treg depletion in young hosts. Anti-Gr-1 antibody did not further augment anti-tumor immunity or tumor control in young mice as denileukin diftitox did not increase their MDSC. Dual Treg plus MDSC depletion increased interferon-γ-producing CD4+ and CD8+ T cells, and
T
CD8+ tumor-specific T cells in aged mice, mirroring the effects on these T cell subsets seen in young mice
IP
treated with Treg depletion alone. To our knowledge this is the first cancer immunotherapy that treats aged, but
CR
not young hosts [5]. Figure 1 outlines the differences in young versus aged mice accounting for differential treatment outcomes. Detailed data on age effects on human T cell subsets in responses to cancer
AN
US
immunotherapy are not yet reported to our knowledge.
M
Age effects of Tregs in human cancer immunotherapy are not yet specifically reported to our knowledge. In
ED
209 melanoma patients receiving αCTLA-4 immunotherapy, high Tregs (and also eosinophils and
PT
lymphocytes) correlated with better clinical outcomes [55], but age was not specifically assessed as a variable.
CE
1.3 OTHER CELLS
Lower T cell function and chronic, low-level age-related inflammation can increase suppressive myeloid cells.
AC
Thus, targeting aged innate cells could improve anti-tumor T cell functions in elderly cancer patients [56].
1.3.1 Dendritic cells
Dendritic cells (DCs) are antigen-presenting cells important in promoting T cell immunity [57]. The local tumor environment favors increasing numbers of dysfunctional DC that hinder anti-tumor immunity and immunotherapy outcomes [58, 59].
6
ACCEPTED MANUSCRIPT DC precursors in blood and Langerhans dendritic cells in skin, conventional DC in other organs and DC generation from bone marrow can all decrease with advancing age, which could contribute to reduced protective immunity with age. By contrast, other DC subsets, notably those with potential autoimmune reactivity can increase in certain animal models of autoimmunity, which could help explain the age-related propensity for
T
autoimmunity, described in detail in [60, 61].
IP
Distinct DC functions with age can be decreased or unchanged depending on the function, such as reduced
CR
alloreactivity in a mixed lymphocyte reaction, reduced T cell activation molecules (e.g., MHC class II, ICAM-1), reduced migration and antigen capture capacity, and reduced cytokine or chemokine production, reviewed
US
extensively in [62]. Some of these effects are DC-intrinsic whereas others (such as reduced migration) could be attributable to defective host factors. If declining or altered DC functions reduce T cell function or promote
AN
inflammation, that could increase age-related cancer risk. Augmenting the capacity of DC to present antigens, a function that can decline with age, induces potent tumor-specific cytotoxic T cell activities. CD40L or agonist
M
anti-CD40 antibodies augment DC activation in human and animal studies [63]. A vaccine consisting of CD40L
ED
hooked to specific cancer antigens showed promise in older cancer patients [64].
PT
In aged humans, reduced Langerhans DC have also been reported [65]. The two principal circulating human
CE
DC subsets (myeloid and plasmacytoid) also appear to decline in numbers and function with age as does DC generation from bone marrow, all effects similar to those seen in aged mice, reviewed in [62]. Interestingly,
AC
although in situ DC from the aged humans have apparent functional defects, when DC are generated from aged precursors (such as monocyte-derived DC), their numbers and functions appear similar to those from young precursors. Thus, DC-based cancer immunotherapies based on generation from precursor cells could be a viable strategy in aged cancer patients. The effects of age on tumor microenvironment-driven DC dysfunction in humans are not reported to our knowledge, but would be useful to know.
7
ACCEPTED MANUSCRIPT 1.3.2 Macrophages Myelopoiesis increases in age [66], leading to increased myeloid cell numbers. Macrophages are important elements of tumor stroma and contribute to tumor-associated immune dysfunction [67]. Tumor macrophages can be pro-inflammatory M1 or anti-inflammatory M2 macrophages, and these phenotypes can interconvert. M1 macrophages produce pro-inflammatory cytokines including TNF-α and IL-12 that augment anti-tumor
T
immunity. M2 macrophages are generally anti-inflammatory and produce cytokines such as IL-10, TGF-β that
CR
IP
promote tumorigenesis or angiogenesis [67].
Macrophages increase in lymphoid organs of aged mice [68]. M1 macrophage functions could increase
US
because of increased age-related reactive oxygen species, but others report reduced M1 macrophage function with age [69], perhaps from IL-10 producing M2 macrophages that increase with age [67]. Tumor-associated
AN
macrophages from aged mice produce high levels of transforming growth factor (TGF)-β that can be immunosuppressive, consistent with an M2-type phenotype, whereas young macrophages do not [68].
M
Detrimental M2 macrophages can be converted to beneficial M1 using IL-12 or CpG plus an αIL-10 receptor
ED
[70]. Thus targeting tumor-associated age-related M2 macrophages could be useful to treat cancers in aged
PT
hosts.
CE
The distribution and some functions of macrophages in aged hosts differs from the young. For example, M1 macrophages with a pro-inflammatory phenotype are increased in liver and fat tissues of normal aged hosts
AC
versus young hosts whereas anti-inflammatory/immune suppressive M2 macrophages are more prevalent in normal aged versus young hosts in peripheral lymphoid tissues, reviewed in [71]. However, effects in cancer, and whether macrophages from aging hosts are especially prone to promote tumor growth in not fully settled and likely will depend on the specific tumor and/or anatomic site.
8
ACCEPTED MANUSCRIPT 1.3.3 Myeloid derived suppressor cells Myeloid derived suppressor cells (MDSC) are immunosuppressive, immature myeloid cells that are elevated in inflammatory diseases including cancers [72-75] and they suppress anti-tumor immunity [76]. MDSCs produce inhibitory molecules (e.g., arginase, IL-10) that inhibit T cell functions, and can promote generation of detrimental M2 macrophages or Tregs [77]. MDSCs increase in blood during human aging [78] and increase
T
in lymphoid organs in mice [5, 68]. MDSC are immunopathologic in age including in aged hosts with cancer
IP
[79, 80]. Treg depletion using denileukin diftitox increased MDSC numbers in aged mice bearing B16
CR
melanomas, suggesting MDSC control by Tregs [5]. In CT26 colon cancer, an extract of Lentinula edodes mycelia reduced MDSC numbers when combined with a cancer vaccine in aged mice. The proposed
US
mechanism was suppression of IL-6 and TNF-α known to promote MDSC [81]. This mycelium extract also improved priming of tumor-specific cytotoxic T cells by the vaccine. Thus, MDSC are an attractive target to
AN
develop potentially effective approaches to mitigate cancer-associated immune dysfunction in the aged host.
M
Age effects of MDSC in human cancer immunotherapy are not yet specifically reported to our knowledge. In
ED
209 patients receiving αCTLA-4 as melanoma immunotherapy, low baseline blood MDSC (and also lactic acid dehydrogenase and monocytes) correlated with better clinical outcomes[55], but age was not independently
CE
1.3.4 B cells
PT
assessed.
AC
Aging B cells can be dysfunctional to normal immune homeostasis by producing tumor necrosis factor-α that is detrimental [82]. We showed that in aged mice aging-associated PD-L2-expressing B cells enhance anti-tumor immunity to ovarian cancer via Th1 and Th17 induction [83]. Little else regarding B cells in anti-tumor immunity or response to immunotherapy has been reported to our knowledge.
9
ACCEPTED MANUSCRIPT 1.3.5 Hematopoietic stem cells As many immune cells come from hematopoietic stem cells, which themselves are subject to aging effects, understanding aging of hematopoietic stem cells could provide insights into improving an aging immune system. Progress on this front has been made [84].
T
1.3.6 Novel age-related immune cells
IP
With age, novel immune cell subsets appear, including those with potential to deviate anti-tumor immunity or
CR
immunotherapy responses. We described PD-L2-expressing B cells that increase with age and enhance antitumor immunity to ovarian cancer via Th1 and Th17 induction. These cells were not seen in young mice [83].
US
CD25lo Tregs appear with age in normal mice [85], in contrast to conventional CD25hi Tregs in young hosts. These are slightly hypofunctional versus CD25hi Tregs but we did not define clear immunopathologic
AN
significance in cancer immunotherapy of aged mice with B16 melanoma although denileukin diftitox reduced
M
their numbers [5].
2.1 Immune checkpoint inhibitors
ED
2.0 SPECIFIC APPROACHES
PT
Immune checkpoint receptor blockade is a highly successful immunotherapy strategy for various cancers [86,
CE
87]. Six distinct checkpoint inhibitor antibody treatments were FDA approved as cancer immunotherapy for a wide variety of cancers at the time of this writing, with more approvals expected soon. Nonetheless, effects in
AC
elderly patients are little reported.
Immune checkpoint molecules control the magnitude of immune responses either positively (activating) or negatively (inhibiting). Immune checkpoint inhibitor antibodies block negative signals from the immune checkpoints elevated in many cancers, and thus can improve anti-cancer immunity [86, 87]. Inhibitory immune checkpoint molecules increase on aged T cells in humans and mice [5, 88, 89], suggesting increased memorylike T cells that also express these molecules and which are hyporesponsive. Immune checkpoint molecules associated with exhausted (poorly functional) T cells, including PD-1, Lag-3 and Tim-3 increase with age as 10
ACCEPTED MANUSCRIPT well. These and other check point molecules are targets for anti-cancer immunotherapies. Other immune checkpoint receptors such as PD-L1 are highly expressed on young myeloid or B cells, but can be at high levels on tumor cells and aged CD8+ T cells [90]. Age-specific strategies to mitigate inhibitory immune checkpoint signals and reduce hyporesponsiveness or exhaustion of aged T cells without compromising anti-
CR
IP
T
tumor effects is an important goal in the development of these modalities.
2.1.1 αPD-1 antibody
US
PD-1 impedes anti-tumor PD-1+ T cells. Monoclonal αPD-1 antibodies (αPD-1) are remarkably effective against many cancers and two distinct αPD-1 antibodies (pembrolizumab and nivolumab) are FDA-approved to
AN
treat melanoma, renal cell carcinoma, non-small cell lung cancer, Hodgkin’s disease, bladder cancer and head
M
and neck cancer.
ED
PD-1 is largely expressed by effector-memory (CD44hiCD62Llo) T cells that are elevated with age. CD4+PD-1+ T cells in old mice are hypoproliferative, suggesting that PD-1 could contribute to the functional decline in
PT
effector-memory T cells with age [91, 92]. Although αPD-1 improves the functioning of T cells in aged mice
CE
[93], this strategy has not been reported as cancer therapy in aged mice to our knowledge. We showed that the mTOR inhibitor rapamycin lowers T cell PD-1 expression with age, and the PD-1+ T cells in rapamycinmice functioned significantly better than PD-1+ T cells in aged mice on control treatment [92],
AC
treated
suggesting that mTOR inhibition could improve anti-tumor immunity in aged hosts. In this regard, everolimus, an mTOR inhibitor similar to rapamycin, improved B cell immune responses to influenza vaccine in aged humans [94].
2.1.2 αPD-L1 antibody PD-L1 is an immune checkpoint molecule that is a major ligand for PD-1. Like αPD-1, αPD-L1 is thought to work as cancer immunotherapy by shielding PD-1+ anti-tumor T cells from tumor PD-L1 inhibition [87, 95-98]. 11
ACCEPTED MANUSCRIPT Three different αPD-L1 antibodies (atezolizumab, avelumab and durvalumab) are now FDA-approved for cancer treatment.
Most old, naive CD8+ T cells in mice are PD-L1+ versus just 25% in young mice. Aged CD8+ T cells exhibited relatively low proliferation, but αPD-L1 increased proliferation in vitro to a level comparable to young CD8+ T
T
cells. αPD-L1 augmented anti-tumor immunity in aged hosts to the level of young hosts in a lymphoma model
CR
IP
in mice [90].
We showed that αPD-L1 treats B16 melanoma in young mice but fails in aged mice. αPD-L1 treatment efficacy
US
was partially restored in combination with αCTLA-4 [99]. The above study [90] tested αPD-L1 in a lymphoma model in Balb/c mice whereas we studied a melanoma in BL6 mice. Thus, either lymphoma versus melanoma,
AN
or host genetic background among other considerations could help explain different results with αPD-L1 in
M
aged hosts.
ED
2.1.3 αCTLA-4 antibody
Ipilimumab, an anti-CTLA-4 antibody, was the first immune checkpoint inhibitor agent approved by the United
PT
States Food and Drug Administration, and the first agent in this class that clearly benefits human cancer by
CE
improving disease-free and overall survival in metastatic melanoma [100]. We showed that αCTLA-4 was the only immunotherapy effective in aged mice as a single agent in B16 melanoma. However, we could not identify
AC
a clear mechanism based on assessments of T cell activation and cytokine production. Further, αCTLA-4 was less able to reduce Tregs specifically in the tumor compared to young tumor-bearing hosts [99]. This potentially important set of data on this agent merits additional investigations.
In 82 melanoma patients receiving αCTLA-4 as immunotherapy, increases in peripheral blood T cells on treatment predicted good outcomes, but the effects did not reach statistical significance[101]. In 209 melanoma patients receiving αCTLA-4 immunotherapy, low baseline blood MDSC, lactic acid dehydrogenase and monocytes, and high Tregs, eosinophils and lymphocytes correlated with better clinical outcomes[55]. Age was 12
ACCEPTED MANUSCRIPT not independently assessed in either of these two studies. It was just reported [102] that in a cohort of 254 melanoma patients receiving αPD-1 or αPD-L1 antibodies as immunotherapy, there was no treatment or survival response difference by age in cohorts of patients aged <50, 50-64, 65-74 or >75 years. However, individual cohorts were small and likely underpowered, thus without clear ability to exclude age as a factor affecting αCTLA-4 efficacy in human melanoma, a finding that we predict will eventually be made.
T
Nonetheless, these are important initial steps in understanding if and how age will alter immune checkpoint
CR
IP
therapy responses as predicted from mouse studies.
2.2 IMMUNE CHECKPOINT AGONISTS
US
Agonists for CD137 and OX40 were described above under T CELLS [30, 33]. These are now in human
AN
clinical trials for cancer, but not with an age focus to our knowledge.
2.3 ADOPTIVE CELL TRANSFERS
M
Adoptive cell transfers of DC or tumor infiltrating lymphocytes were generally poorly effective against cancers.
ED
Transfers of chimeric antigen receptor (CAR) cells show greater promise in hematologic malignancies. To make cell expressing a CAR, the CAR is genetically engineered into lymphocytes that are then transfused into
PT
the patient [103]. We are unaware of published reports of age effects on immune cells in generating CAR cells,
CE
aside from understanding that older T cells generally yield fewer CAR T cells versus younger patients following in vitro culture. If aged T cells have reduced cytolytic capacity or homing, they could be less effective as CAR T
AC
cell cancer immunotherapy. Means to boost aged T cell functions discussed above could potentially boost of CAR T cell efficacy from aged T cells. Natural killer cells and γδ T cells do not require a major histocompatibility match to kill targets cells. Thus, CAR engineered into these cells might be useful to treat aged cancer patients, as CAR NK and γδ T cells are under investigation. Similarly, aged DC with reduced T cell activating or antigen presenting capacities, could limit efficacy when made form aged hosts, although strategies to boost aged DC functions as discussed above could be evaluated.
13
ACCEPTED MANUSCRIPT 2.4 mTOR INHIBITORS Mammalian target of rapamycin (mTOR) inhibitors at doses lower than those to induce immunosuppression improve antigen-specific immunity, including tumor-specific immunity [104]. We reported that in aged mice, rapamycin reduces their T cell PD-1 expression and improves their PD-1+ T cell functions [47]. Chronic administration of rapamycin in the eRapa microencapsulated formulation had major effects on many T cell
T
functions including increasing IL-17 and reducing interferon-γ production, altering Th (CD4+ T helper)
IP
polarization in many Th pathways but with little effect on Treg numbers, altering T cell trafficking molecules,
CR
altering many canonical regulatory pathways, reducing total and memory T cells and had many other significant T cell effects in diverse organs [47]. Surprisingly, effects in all these diverse outcomes were
AN
US
generally similar in young and aged hosts and in males and females.
T cell differentiation depends on mTOR, and mTOR suppression can promote Treg function or differentiation
M
[92, 105]. Thus, small molecule mTOR inhibitors could blunt anti-tumor immunity by generating detrimental
ED
Tregs, as Tregs can inhibit anti-tumor immunity [37]. However, in the right settings and at the appropriately low dose, small molecule mTOR inhibitors do yield net anti-tumor immunity benefits [47, 82, 92, 93] and do not
PT
appear to promote increased Treg numbers or function [92]. We reported that low dose rapamycin is beneficial
CE
to anti-cancer immunotherapy whereas typical therapeutic rapamycin doses impair it [106].
AC
Chronic eRapa administration also increased B cell prevalence in spleen and bone marrow of young and aged mice but without significant change in absolute numbers. Follicular and marginal zone B cells were increased and transitional 1 (T1) B cells were decreased. Phenotyping and genomic data suggested that eRapa preserved naıve B cells. Strikingly, eRapa was not consistently anti-inflammatory as expected, but induced or inhibited distinct inflammatory pathways. eRapa appeared to increase numbers and functions on innate lymphoid cells as evidenced by their IL-17 and especially IL-22 production, either of which could be pro-tumor or tumor-protective in distinct settings. Diverse effects on myeloid cells and natural killer cells were also
14
ACCEPTED MANUSCRIPT observed. As noted in T cell studies, eRapa effects in all of the distinct cell types, sexes and anatomic compartments were generally similar in young and aged mice [47],
We further found that rapamycin improves γδ T cell-mediated anti-cancer immunity [107]. Thus, mTOR inhibitors might be useful as adjuncts to cancer immunotherapy agents in aged patients, as we showed is
T
possible [92] (and see related article in this issue). Rapamycin prolongs life of mice even when given late in
IP
life [108] which could be due to its cancer prevention effects. However, we unexpectedly found that eRapa
CR
significantly prolonged life in highly immune deficient mice lacking critical anti-cancer immune defenses [47].
US
More more work on mTOR effects is needed.
AN
2.5 CALORIC RESTRICTION
Similar to rapamycin, caloric restriction suppresses mTOR. Caloric restriction boosted αCD40 treatment of
M
breast cancer and sarcoma when used over 6-8 months in mice that were 12 months old. Priming of tumor
ED
antigen-specific CD4+ but not CD8+ T cells was similar to that seen in young controls [109]. Certain approaches to caloric restriction are tolerated by aged patients, and can improve anti-tumor immunity in young
PT
subjects [110]. However, mTOR suppression can also reduce immunity in aged hosts [111] and at too late an
CE
age or at too great a caloric reduction, caloric restriction harms aged hosts. Caloric restriction merits further
AC
studies to mitigate cancer treatment symptoms, or prevent or treat cancer in aged hosts.
2.6 TOLL-LIKE RECEPTOR AGONISTS Bacille Calmette-Guérin (BCG), an attenuated Mycobacterium bovis is approved by the United States Food and Drug Administration for non-muscle invasive bladder cancer. It potently agonizes Toll like receptors. Although its mechanism of anti-cancer action is not fully understood, it is thought to improve anti-cancer immunity. Observational studies suggest that age influences BCG therapy responses in bladder cancer. Aged bladder cancer patients had increased relapse rates following BCG compared to younger patients in a trial of BCG plus interferon-α immunotherapy [112]. In patients aged 61-70 years old versus those older than 80, 15
ACCEPTED MANUSCRIPT cancer-free survival was 39% versus 61%, respectively, at median follow-up of 24 months. Age was an independent risk for progression in aged bladder cancer patients treated with BCG, and the 2-year progression-free survival was 87% for patients under 75 years versus 65% in patients older than 75 years [113]. In a large cohort of 805 patients with bladder cancer, age had a small (but measurable) effect on reducing BCG therapy response [114]. However, selective surgical management of elderly patients versus
T
immune effects and competing morbidities effects are not clear from these studies. Thus, BCG is the only
IP
agent of which we are aware for which age-specific human immunotherapy effects are extensively reported.
CR
Nonetheless, firm age-related immune effect conclusions cannot yet be made. As BCG is standard of care immunotherapy and as bladder cancer patients are typically older than for many other cancer types, additional
US
studies are warranted.
AN
3.0 TUMOR MICROENVIRONMENT
The tumor microenvironment includes the tumor itself, infiltrating immune cells, stromal cells and matrix The
aged
tumor
microenvironment
M
components.
has
aspects
that
suggest
it
could
be
more
ED
immunosuppressive versus in young hosts [115]. Such aspects include increased M2 macrophages and MDSC that could be attributable to chronic, low grade age-related inflammation. Other factors include tumor
PT
cells that can attract detrimental MDSC, neutrophils or Tregs that blunt anti-tumor immunity and can generate
CE
pro-tumorigenic molecules [35, 68]. For example, stromal fibroblasts from older versus younger prostate
AC
cancer patients generate more pro-inflammatory factors [116].
In a transgenic prostate cancer model and model of benign prostate hyperplasia in mice, there was an agerelated increase in TGF-β1 that further altered local tissue structure suggesting local inflammation [117]. Data aged prostates in normal mice show similar effects [118]. Aged fibroblasts attract CD4+ T cells that support proliferation of prostate epithelial cells. Other cells, including macrophages, CD8+ T cells and neutrophils promote prostate cancer cell proliferation [90].
16
ACCEPTED MANUSCRIPT In the microenvironment of aged melanoma, fibroblasts promote melanoma growth and progression. Senescence in aged fibroblasts could drive tumor metastases and angiogenesis by producing frizzled-related protein 2. Older fibroblasts also generate fewer scavengers of reactive oxygen species that can in turn promote more DNA damage [6]. Aged mice receiving agonist αCD40 did not to respond to treatment because of defective CD40 tumor microenvironmental signals, not due to T cell-intrinsic effects [32]. Thus, treatments that
T
modify the stromal, could be relevant for elderly cancer patients, an area of investigation that has not seen
CR
IP
much reported research activity to date.
4.0 IMMUNOTHERAPY TOXICITY MITIGATION
US
In aged mice, cancer immunotherapy can be lethal or produce local toxicities or toxicities in distinct organs, in part due to pro-inflammatory cytokines that are generated locally in affected tissues and systemically.
AN
Macrophages appear to be prime drivers of pro-inflammatory effects over T cells or natural killer cells. Depletion of myeloid cells concomitantly with therapy can mitigate toxicities and yet not compromise treatment
M
efficacy significantly. Aged macrophages from mice and from normal humans generally produce higher levels
ED
of pro-inflammatory cytokines, including TNF-α and IL-6 versus younger macrophages. TNF-α blockade improves survival and reduces toxicities without loss of anti-tumor effects in aged mice [3, 4]. αTNF-α, αIL-6
PT
and αIL-6R antibodies are all FDA-approved. Thus, this concept merits further investigation for potential rapid
CE
translation as justified by human data. However, administration of IL-6 and TNF-α administration can improve aged T cell functions [29]. Thus, much additional work is needed to use such approaches safely and effectively
AC
in cancer immunotherapy strategies.
5.0 MICROBIOME EFFECTS Gut microbes affects systemic immunity and the gut microbial composition is altered by age [119-121]. In two separate papers gut microbes were shown to affect anti-cancer immunotherapy in human subjects being treated with αPD-L1 [122] or αCTLA-4 [123]. At present, there is much justified interest in defining microbial effects on anti-cancer immunity. Age effects in this regard are not yet reported on cancer immunotherapy to our knowledge. As gut microbes can affect anti-cancer treatment outcomes by modulating immunity [124], 17
ACCEPTED MANUSCRIPT including modulating tumor microenvironmental myeloid cells [125], themselves affected with age [92], it is predictable that we will identify age-associated microbial effects on anti-cancer immunotherapy, an area meriting further work.
6.0 CONCLUSIONS
T
Cancer immunotherapy has a strong scientific rationale. Recent advances in understanding details of cancer
IP
driven immune dysfunction have helped develop highly successful, although still imperfect, anti-cancer
CR
immunotherapies. Much of our understanding of anti-tumor immunity and responses to cancer immunotherapy derives from studies of relatively young hosts. A growing, but still relatively small, body of data is beginning to
US
define age effects on cancer immunity and responses to immunotherapies. There are some examples of novel approaches to improving anti-cancer immunotherapy in aged hosts most at risk for cancer and in means to
AN
mitigate toxicities of immunotherapy with emphasis on age effects.
M
“Immune decline” is an inaccurate term in reference to the aged immune system, and “immunosenescence”
ED
and “inflammaging” describe specific attributes of an aged immune system. We propose the term “Age Related Immune Dysfunction” (ARID) to describe the totality of age-related immune changes to include increased or
PT
decreased numbers of certain immune cells, appearance of novel immune cell populations, increased pro-
CE
inflammatory or immune suppressive functions, restricted TCR repertoire and the myriad other changes that age brings to immunity. To the extent that underlying ARID can be reversed in aged cancer patients, clinically
AC
relevant anti-tumor immunity could potentially be achieved as has now clearly been demonstrated.
7.0 CHALLENGES IN DEVELOPING EFFECTIVE AGE-APPROPRIATE CANCER IMMUNOTHERAPY Understanding age effects on tumor-specific immunopathology and related age-specific effects better will help in development of more effective treatments. Individual agents are unlikely to be effective cancer treatments, and thus means to combine agents and approaches optimally needs further definition, including age-specific agents, doses and schedules. Biomarkers that differentiate aged patients with potential to generate tumor18
ACCEPTED MANUSCRIPT specific immunity to selected agents from those patients requiring alternative or adjunctive approaches are also needed. Adjuncts could include adoptive transfer of young or rejuvenated CAR-expressing immune cells, improved antigen presenting cells, thymic transplants, gene therapy to improve specific age-associated defects, microbial enhancements or other approaches.
T
Toxicity mitigation strategies that do not negate clinical efficacy are needed, as is means to identify rational
IP
therapy combinations that minimize use of potentially harmful individual agents while preserving treatment
CR
efficacy. Studies of age effects of the human gut microbiome are early, but data from them could help understand heterogeneous immunotherapy responses in humans and in pre-clinical models. Aside from the
US
gut microbiome, age effects on other microbial populations (e.g., lung, skin) require additional attention. The mutational burden of individual cancers affects efficacy of cancer immunotherapies. This mutational burden is
AN
dictated by immune editing, among other factors. Thus, we must understand age effects on immune editing and tumor mutations, which information could improve cancer immunotherapy efficacy. Finally, we must
M
deepen our knowledge of age effects on tumor stroma, including vasculature and matrix effects that could
ED
affect immune cell or drug infiltration. As ever more powerful but costly and potentially harmful approaches are brought to bear on ever older cancer patients, economic, feasibility and ethical issues must be addressed in
CE
PT
greater depth.
Notable similarities in mouse models of the aging immune system with the human immune system include a
AC
general reduction in important immune cells including T cells and DC, increase in memory phenotype immune cells, reduction in naïve phenotype immune cells, increased MDSC and general increase in inflammatory markers. The ability to respond to vaccines is generally impaired in aged mice and humans. There is little data on effects of age on human responses to cancer immunotherapy including T cell subset-specific data, effects on induction of tumor-specific immune memory, effects of T cell exhaustion and effects of Tregs, MDSC and other immunosuppressive cells and factors on immune and clinical outcomes. The single report thus far on age effects in immune checkpoint blockade in melanoma immunotherapy did not demonstrate an age effect on clinical outcomes, but this study could have been underpowered. Mouse models will be useful to test certain 19
ACCEPTED MANUSCRIPT hypotheses and strategies in approaches to cancer immunotherapy in aged hosts, but it is clear tat much additional human data is also needed to address this problem that we consider to be important, but about which many important top-pevel questions remain unaddressed in detail.
Acknowledgements: VH, RSS and TJC wrote the manuscript. VH and AP performed some experiments
IP
T
described.
AC
CE
PT
ED
M
AN
US
CR
Conflict of Interest: The authors declare no financial conflicts.
20
ACCEPTED MANUSCRIPT LITERATURE CITED 1.
Pardoll D. T cells and tumours. Nature 2001; 411:1010-2.
2.
Pardoll D. Does the immune system see tumors as foreign or self? Annu Rev Immunol 2003; 21:807-39.
3.
Bouchlaka MN, Murphy WJ. Impact of aging in cancer immunotherapy: The importance of using accurate preclinical models. Oncoimmunology 2013; 2:e27186. Bouchlaka MN, Sckisel GD, Chen M, Mirsoian A, Zamora AE, Maverakis E, Wilkins DE, Alderson KL, Hsiao HH,
T
4.
IP
Weiss JM, Monjazeb AM, Hesdorffer C, Ferrucci L, Longo DL, Blazar BR, Wiltrout RH, Redelman D, Taub DD,
CR
Murphy WJ. Aging predisposes to acute inflammatory induced pathology after tumor immunotherapy. J Exp Med 2013; 210:2223-37.
Hurez V, Daniel BJ, Sun L, Liu AJ, Ludwig SM, Kious MJ, Thibodeaux SR, Pandeswara S, Murthy K, Livi CB, Wall S,
US
5.
AN
Brumlik MJ, Shin T, Zhang B, Curiel TJ. Mitigating age-related immune dysfunction heightens the efficacy of tumor immunotherapy in aged mice. Cancer Res 2012; 72:2089-99. Kaur A, Webster MR, Marchbank K, Behera R, Ndoye A, Kugel CH, 3rd, Dang VM, Appleton J, O'Connell MP,
M
6.
ED
Cheng P, Valiga AA, Morissette R, McDonnell NB, Ferrucci L, Kossenkov AV, Meeth K, Tang HY, Yin X, Wood WH, 3rd, Lehrmann E, Becker KG, Flaherty KT, Frederick DT, Wargo JA, Cooper ZA, Tetzlaff MT, Hudgens C, Aird KM,
PT
Zhang R, Xu X, Liu Q, Bartlett E, Karakousis G, Eroglu Z, Lo RS, Chan M, Menzies AM, Long GV, Johnson DB,
CE
Sosman J, Schilling B, Schadendorf D, Speicher DW, Bosenberg M, Ribas A, Weeraratna AT. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature 2016; 532:250-4. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and
AC
7.
promotion. Science 2011; 331:1565-70. 8.
Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol 2004; 22:329-60.
9.
Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, Smyth MJ, Schreiber RD. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 2007; 450:903-7.
21
ACCEPTED MANUSCRIPT 10.
Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK, Hundal J, Wendl MC, Demeter R, Wylie T, Allison JP, Smyth MJ, Old LJ, Mardis ER, Schreiber RD. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012; 482:400-4.
11.
DuPage M, Mazumdar C, Schmidt LM, Cheung AF, Jacks T. Expression of tumour-specific antigens underlies cancer immunoediting. Nature 2012; 482:405-9. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component
T
12.
Strauss DC, Thomas JM. Transmission of donor melanoma by organ transplantation. Lancet Oncol 2010; 11:790-
CR
13.
IP
phases--elimination, equilibrium and escape. Curr Opin Immunol 2014; 27:16-25.
6.
Stephens JK, Everson GT, Elliott CL, Kam I, Wachs M, Haney J, Bartlett ST, Franklin WA. Fatal transfer of
US
14.
AN
malignant melanoma from multiorgan donor to four allograft recipients. Transplantation 2000; 70:232-6. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100:57-70.
16.
Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer
M
15.
17.
ED
Cell 2012; 21:297-308.
Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, Trinchieri G, Dubinett SM, Mao JT, Szabo E,
PT
Krieg A, Weiner GJ, Fox BA, Coukos G, Wang E, Abraham RT, Carbone M, Lotze MT. Cancer and inflammation:
CE
promise for biologic therapy. J Immunother 2010; 33:335-51. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140:883-99.
19.
Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of
AC
18.
cancer: links to genetic instability. Carcinogenesis 2009; 30:1073-81. 20.
Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, Savage DA, Mueller-Holzner E, Marth C, Kocjan G, Gayther SA, Jones A, Beck S, Wagner W, Laird PW, Jacobs IJ, Widschwendter M. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res 2010; 20:440-6.
22
ACCEPTED MANUSCRIPT 21.
Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability--an evolving hallmark of cancer. Nat Rev Mol Cell Biol 2010; 11:220-8.
22.
De Bock K, Cauwenberghs S, Carmeliet P. Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr Opin Genet Dev 2010; 21:73-9. Hakim FT, Flomerfelt FA, Boyiadzis M, Gress RE. Aging, immunity and cancer. Curr Opin Immunol 2004; 16:151-6.
24.
Haynes BF, Sempowski GD, Wells AF, Hale LP. The human thymus during aging. Immunol Res 2000; 22:253-61.
25.
Surh CD, Boyman O, Purton JF, Sprent J. Homeostasis of memory T cells. Immunol Rev 2006; 211:154-63.
26.
Rossi DJ, Bryder D, Zahn JM, Ahlenius H, Sonu R, Wagers AJ, Weissman IL. Cell intrinsic alterations underlie
CR
IP
T
23.
hematopoietic stem cell aging. Proc Natl Acad Sci U S A 2005; 102:9194-9.
Fulop T, Larbi A, Pawelec G. Human T cell aging and the impact of persistent viral infections. Front Immunol
US
27.
28.
AN
2013; 4:271.
Fulop T, Witkowski JM, Le Page A, Fortin C, Pawelec G, Larbi A. Intracellular signalling pathways: targets to
Haynes L, Eaton SM, Burns EM, Rincon M, Swain SL. Inflammatory cytokines overcome age-related defects in
ED
29.
M
reverse immunosenescence. Clin Exp Immunol 2016.
CD4 T cell responses in vivo. J Immunol 2004; 172:5194-9. Bansal-Pakala P, Croft M. Defective T cell priming associated with aging can be rescued by signaling through 4-
PT
30.
31.
CE
1BB (CD137). J Immunol 2002; 169:5005-9. Ruby CE, Weinberg AD. OX40-enhanced tumor rejection and effector T cell differentiation decreases with age. J
32.
AC
Immunol 2009; 182:1481-9.
Ruby CE, Weinberg AD. The effect of aging on OX40 agonist-mediated cancer immunotherapy. Cancer Immunol Immunother 2009; 58:1941-7.
33.
Lustgarten J, Dominguez AL, Thoman M. Aged mice develop protective antitumor immune responses with appropriate costimulation. J Immunol 2004; 173:4510-5.
23
ACCEPTED MANUSCRIPT 34.
Wang W, Kryczek I, Dostal L, Lin H, Tan L, Zhao L, Lu F, Wei S, Maj T, Peng D, He G, Vatan L, Szeliga W, Kuick R, Kotarski J, Tarkowski R, Dou Y, Rattan R, Munkarah A, Liu JR, Zou W. Effector T Cells Abrogate Stroma-Mediated Chemoresistance in Ovarian Cancer. Cell 2016; 165:1092-105.
35.
Jackaman C, Nelson DJ. Are macrophages, myeloid derived suppressor cells and neutrophils mediators of local suppression in healthy and cancerous tissues in aging hosts? Exp Gerontol 2014; 54:53-7. Curiel TJ. Tregs and rethinking cancer immunotherapy. J Clin Invest 2007; 117:1167-74.
37.
Curiel TJ. Regulatory T cells and treatment of cancer. Curr Opin Immunol 2008; 20:241-6.
38.
Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow
CR
IP
T
36.
M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W. Specific
US
recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival.
AN
Nat Med 2004; 10:942-9.
Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol 2006; 6:295-307.
40.
Kozlowska E, Biernacka M, Ciechomska M, Drela N. Age-related changes in the occurrence and characteristics of
M
39.
41.
ED
thymic CD4(+) CD25(+) T cells in mice. Immunology 2007; 122:445-53. Thomas DC, Mellanby RJ, Phillips JM, Cooke A. An early age-related increase in the frequency of CD4+ Foxp3+
Zhang H, Podojil JR, Luo X, Miller SD. Intrinsic and induced regulation of the age-associated onset of
CE
42.
PT
cells in BDC2.5NOD mice. Immunology 2007; 121:565-76.
spontaneous experimental autoimmune encephalomyelitis. J Immunol 2008; 181:4638-47. Rosenkranz D, Weyer S, Tolosa E, Gaenslen A, Berg D, Leyhe T, Gasser T, Stoltze L. Higher frequency of
AC
43.
regulatory T cells in the elderly and increased suppressive activity in neurodegeneration. J Neuroimmunol 2007; 188:117-27. 44.
Zhao L, Sun L, Wang H, Ma H, Liu G, Zhao Y. Changes of CD4+CD25+Foxp3+ regulatory T cells in aged Balb/c mice. J Leukoc Biol 2007; 81:1386-94.
24
ACCEPTED MANUSCRIPT 45.
Kryczek I, Liu R, Wang G, Wu K, Shu X, Szeliga W, Vatan L, Finlayson E, Huang E, Simeone D, Redman B, Welling TH, Chang A, Zou W. FOXP3 defines regulatory T cells in human tumor and autoimmune disease. Cancer Research 2009; 69:3995-4000.
46.
Sun L, Hurez VJ, Thibodeaux SR, Kious MJ, Liu A, Lin P, Murthy K, Pandeswara S, Shin T, Curiel TJ. Aged regulatory T cells protect from autoimmune inflammation despite reduced STAT3 activation and decreased constraint of IL-
Lages CS, Suffia I, Velilla PA, Huang B, Warshaw G, Hildeman DA, Belkaid Y, Chougnet C. Functional regulatory T
IP
47.
T
17 producing T cells. Aging Cell 2012; 11:509-19.
CR
cells accumulate in aged hosts and promote chronic infectious disease reactivation. J Immunol 2008; 181:183548.
Tsaknaridis L, Spencer L, Culbertson N, Hicks K, LaTocha D, Chou YK, Whitham RH, Bakke A, Jones RE, Offner H,
US
48.
AN
Bourdette DN, Vandenbark AA. Functional assay for human CD4+CD25+ Treg cells reveals an age-dependent loss of suppressive activity. J Neurosci Res 2003; 74:296-308.
Garg SK, Delaney C, Toubai T, Ghosh A, Reddy P, Banerjee R, Yung R. Aging is associated with increased
M
49.
ED
regulatory T-cell function. Aging Cell 2014; 13:441-8. Fessler J, Ficjan A, Duftner C, Dejaco C. The impact of aging on regulatory T-cells. Front Immunol 2013; 4:231.
51.
Gregg R, Smith CM, Clark FJ, Dunnion D, Khan N, Chakraverty R, Nayak L, Moss PA. The number of human
PT
50.
CE
peripheral blood CD4+ CD25high regulatory T cells increases with age. Clin Exp Immunol 2005; 140:540-6. Hwang KA, Kim HR, Kang I. Aging and human CD4(+) regulatory T cells. Mech Ageing Dev 2009; 130:509-17.
53.
Dominguez AL, Lustgarten J. Implications of aging and self-tolerance on the generation of immune and
AC
52.
antitumor immune responses. Cancer Res 2008; 68:5423-31. 54.
Sharma S, Dominguez AL, Lustgarten J. High accumulation of T regulatory cells prevents the activation of immune responses in aged animals. J Immunol 2006; 177:8348-55.
55.
Martens A, Wistuba-Hamprecht K, Geukes Foppen M, Yuan J, Postow MA, Wong P, Romano E, Khammari A, Dreno B, Capone M, Ascierto PA, Di Giacomo AM, Maio M, Schilling B, Sucker A, Schadendorf D, Hassel JC, Eigentler TK, Martus P, Wolchok JD, Blank C, Pawelec G, Garbe C, Weide B. Baseline Peripheral Blood Biomarkers 25
ACCEPTED MANUSCRIPT Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22:2908-18. 56.
Jackaman C, Dye DE, Nelson DJ. IL-2/CD40-activated macrophages rescue age and tumor-induced T cell dysfunction in elderly mice. Age (Dordr) 2014; 36:9655. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265-77.
58.
Zou W, Machelon V, Coulomb-L'Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I,
T
57.
IP
Gordon A, Pustilnik T, Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived factor-1 in human
59.
CR
tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med 2001; 7:1339-46. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC,
US
David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT,
AN
Chen L, Zou W. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 2003; 9:562-7.
Ishikawa S, Sato T, Abe M, Nagai S, Onai N, Yoneyama H, Zhang Y, Suzuki T, Hashimoto S, Shirai T, Lipp M,
M
60.
ED
Matsushima K. Aberrant high expression of B lymphocyte chemokine (BLC/CXCL13) by C11b+CD11c+ dendritic cells in murine lupus and preferential chemotaxis of B1 cells towards BLC. J Exp Med 2001; 193:1393-402. Ishikawa S, Nagai S, Sato T, Akadegawa K, Yoneyama H, Zhang YY, Onai N, Matsushima K. Increased circulating
PT
61.
CE
CD11b+CD11c+ dendritic cells (DC) in aged BWF1 mice which can be matured by TNF-alpha into BLC/CXCL13producing DC. Eur J Immunol 2002; 32:1881-7. Agrawal A, Agrawal S, Tay J, Gupta S. Biology of dendritic cells in aging. J Clin Immunol 2008; 28:14-20.
63.
Khong A, Nelson DJ, Nowak AK, Lake RA, Robinson BW. The use of agonistic anti-CD40 therapy in treatments for
AC
62.
cancer. Int Rev Immunol 2012; 31:246-66. 64.
Tang YC, Thoman M, Linton PJ, Deisseroth A. Use of CD40L immunoconjugates to overcome the defective immune response to vaccines for infections and cancer in the aged. Cancer Immunol Immunother 2009; 58:1949-57.
26
ACCEPTED MANUSCRIPT 65.
Ghersetich I, Lotti T. alpha-Interferon cream restores decreased levels of Langerhans/indeterminate (CD1a+) cells in aged and PUVA-treated skin. Skin Pharmacol 1994; 7:118-20.
66.
Geiger H, Rudolph KL. Aging in the lympho-hematopoietic stem cell compartment. Trends Immunol 2009; 30:360-5. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 2008; 454:436-44.
68.
Jackaman C, Radley-Crabb HG, Soffe Z, Shavlakadze T, Grounds MD, Nelson DJ. Targeting macrophages rescues
T
67.
Mahbub S, Deburghgraeve CR, Kovacs EJ. Advanced age impairs macrophage polarization. J Interferon Cytokine
CR
69.
IP
age-related immune deficiencies in C57BL/6J geriatric mice. Aging Cell 2013; 12:345-57.
Res 2012; 32:18-26.
Watkins SK, Egilmez NK, Suttles J, Stout RD. IL-12 rapidly alters the functional profile of tumor-associated and
US
70.
71.
AN
tumor-infiltrating macrophages in vitro and in vivo. J Immunol 2007; 178:1357-62. Jackaman C, Tomay F, Duong L, Abdol Razak NB, Pixley FJ, Metharom P, Nelson DJ. Aging and cancer: The role of
Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol
ED
72.
M
macrophages and neutrophils. Ageing Res Rev 2017; 36:105-16.
2009; 182:4499-506.
Li H, Han Y, Guo Q, Zhang M, Cao X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells
PT
73.
74.
CE
through membrane-bound TGF-beta 1. J Immunol 2009; 182:240-9. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice.
75.
AC
J Immunol 2008; 181:5791-802.
Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev 2008; 222:162-79.
76.
Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, Divino CM, Chen SH. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumorbearing host. Cancer Res 2006; 66:1123-31.
27
ACCEPTED MANUSCRIPT 77.
Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest 2015; 125:3356-64.
78.
Verschoor CP, Johnstone J, Millar J, Dorrington MG, Habibagahi M, Lelic A, Loeb M, Bramson JL, Bowdish DM. Blood CD33(+)HLA-DR(-) myeloid-derived suppressor cells are increased with age and a history of cancer. J Leukoc Biol 2013; 93:633-7. Grizzle WE, Xu X, Zhang S, Stockard CR, Liu C, Yu S, Wang J, Mountz JD, Zhang HG. Age-related increase of tumor
T
79.
80.
CR
recombinant inbred BXD12 mice. Mech Ageing Dev 2007; 128:672-80.
Enioutina EY, Bareyan D, Daynes RA. A role for immature myeloid cells in immune senescence. J Immunol 2011;
US
186:697-707.
Ishikawa S, Matsui Y, Wachi S, Yamaguchi H, Harashima N, Harada M. Age-associated impairment of antitumor
AN
81.
IP
susceptibility is associated with myeloid-derived suppressor cell mediated suppression of T cell cytotoxicity in
immunity in carcinoma-bearing mice and restoration by oral administration of Lentinula edodes mycelia extract.
Ratliff M, Alter S, McAvoy K, Frasca D, Wright JA, Zinkel SS, Khan WN, Blomberg BB, Riley RL. In aged mice, low
ED
82.
M
Cancer Immunol Immunother 2016; 65:961-72.
surrogate light chain promotes pro-B-cell apoptotic resistance, compromises the PreBCR checkpoint, and favors
Tomihara K, Shin T, Hurez VJ, Yagita H, Pardoll DM, Zhang B, Curiel TJ, Shin T. Aging-associated B7-DC(+) B cells
CE
83.
PT
generation of autoreactive, phosphorylcholine-specific B cells. Aging Cell 2015; 14:382-90.
enhance anti-tumor immunity via Th1 and Th17 induction. Aging Cell 2012; 11:128-38. Florian MC, Nattamai KJ, Dorr K, Marka G, Uberle B, Vas V, Eckl C, Andra I, Schiemann M, Oostendorp RA,
AC
84.
Scharffetter-Kochanek K, Kestler HA, Zheng Y, Geiger H. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 2013; 503:392-6. 85.
Nishioka T, Shimizu J, Iida R, Yamazaki S, Sakaguchi S. CD4+CD25+Foxp3+ T cells and CD4+CD25-Foxp3+ T cells in aged mice. J Immunol 2006; 176:6586-93.
86.
Pardoll D, Drake C. Immunotherapy earns its spot in the ranks of cancer therapy. J Exp Med 2012; 209:201-9.
87.
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12:252-64. 28
ACCEPTED MANUSCRIPT 88.
Canaday DH, Parker KE, Aung H, Chen HE, Nunez-Medina D, Burant CJ. Age-dependent changes in the expression of regulatory cell surface ligands in activated human T-cells. BMC Immunol 2013; 14:45.
89.
Channappanavar R, Twardy BS, Krishna P, Suvas S. Advancing age leads to predominance of inhibitory receptor expressing CD4 T cells. Mech Ageing Dev 2009; 130:709-12.
90.
Mirza N, Duque MA, Dominguez AL, Schrum AG, Dong H, Lustgarten J. B7-H1 expression on old CD8+ T cells
Shimada Y, Hayashi M, Nagasaka Y, Ohno-Iwashita Y, Inomata M. Age-associated up-regulation of a negative co-
IP
91.
T
negatively regulates the activation of immune responses in aged animals. J Immunol 2010; 184:5466-74.
92.
CR
stimulatory receptor PD-1 in mouse CD4+ T cells. Exp Gerontol 2009; 44:517-22.
Hurez V, Dao V, Liu A, Pandeswara S, Gelfond J, Sun L, Bergman M, Orihuela CJ, Galvan V, Padron A, Drerup J, Liu
US
Y, Hasty P, Sharp ZD, Curiel TJ. Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate
93.
AN
lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell 2015; 14:945-56. Lages CS, Lewkowich I, Sproles A, Wills-Karp M, Chougnet C. Partial restoration of T cell function in aged mice by
Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, Lonetto MA, Maecker HT, Kovarik J,
ED
94.
M
in vitro blockade of the PD-1/PD-L1 pathway. Aging Cell 2010.
Carson S, Glass DJ, Klickstein LB. mTOR inhibition improves immune function in the elderly. Sci Transl Med 2014;
Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA,
CE
95.
PT
6:268ra179.
Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA,
AC
Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366:2443-54. 96.
Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat Med 2002; 8:793-800.
29
ACCEPTED MANUSCRIPT 97.
Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455-65.
98.
Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, Chen L.
T
Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an
Figueiredo ASP, Hurez V, Liu A, 2016 TJCJI. Age and sex affect αCTLA-4 efficacy alone and combined with αB7-H1
CR
99.
IP
adaptive resistance mechanism of immune escape. Sci Transl Med 2012; 4:127ra37.
or regulatory T cell depletion in a melanoma model. Journal of Immunology 2016; 196:213.4. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D,
US
100.
AN
Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ.
Martens A, Wistuba-Hamprecht K, Yuan J, Postow MA, Wong P, Capone M, Madonna G, Khammari A, Schilling B,
ED
101.
M
Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711-23.
Sucker A, Schadendorf D, Martus P, Dreno B, Ascierto PA, Wolchok JD, Pawelec G, Garbe C, Weide B. Increases in
PT
Absolute Lymphocytes and Circulating CD4+ and CD8+ T Cells Are Associated with Positive Clinical Outcome of
102.
CE
Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22:4848-58. Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson
103.
June CH, Riddell SR, Schumacher TN. Adoptive cellular therapy: a race to the finish line. Sci Transl Med 2015; 7:280ps7.
104.
AC
DB, Sullivan RJ. Impact of Age on Outcomes with Immunotherapy for Patients with Melanoma. Oncologist 2017.
Pedicord VA, Cross JR, Montalvo-Ortiz W, Miller ML, Allison JP. Friends not foes: CTLA-4 blockade and mTOR inhibition cooperate during CD8+ T cell priming to promote memory formation and metabolic readiness. J Immunol 2015; 194:2089-98.
30
ACCEPTED MANUSCRIPT 105.
Pollizzi KN, Patel CH, Sun IH, Oh MH, Waickman AT, Wen J, Delgoffe GM, Powell JD. mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J Clin Invest 2015; 125:2090-108.
106.
Liu Y, Pandeswara S, Dao V, Padron A, Drerup JM, Lao S, Liu A, Hurez V, Curiel TJ. Biphasic Rapamycin Effects in Lymphoma and Carcinoma Treatment. Cancer Res 2017; 77:520-31.
107.
Dao V, Liu Y, Pandeswara S, Svatek RS, Gelfond JA, Liu A, Hurez V, Curiel TJ. Immune-Stimulatory Effects of
Hasty P, Livi CB, Dodds SG, Jones D, Strong R, Javors M, Fischer KE, Sloane L, Murthy K, Hubbard G, Sun L, Hurez
IP
108.
T
Rapamycin Are Mediated by Stimulation of Antitumor gammadelta T Cells. Cancer Res 2016; 76:5970-82.
CR
V, Curiel TJ, Sharp ZD. eRapa Restores a Normal Life Span in a FAP Mouse Model. Cancer Prev Res (Phila) 2014; 7:169-78.
Farazi M, Nguyen J, Goldufsky J, Linnane S, Lukaesko L, Weinberg AD, Ruby CE. Caloric restriction maintains
US
109.
AN
OX40 agonist-mediated tumor immunity and CD4 T cell priming during aging. Cancer Immunol Immunother 2014; 63:615-26.
Di Biase S, Lee C, Brandhorst S, Manes B, Buono R, Cheng CW, Cacciottolo M, Martin-Montalvo A, de Cabo R,
M
110.
ED
Wei M, Morgan TE, Longo VD. Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Cancer Cell 2016; 30:136-46.
Goldberg EL, Romero-Aleshire MJ, Renkema KR, Ventevogel MS, Chew WM, Uhrlaub JL, Smithey MJ, Limesand
PT
111.
CE
KH, Sempowski GD, Brooks HL, Nikolich-Zugich J. Lifespan-extending caloric restriction or mTOR inhibition impair adaptive immunity of old mice by distinct mechanisms. Aging Cell 2014. Joudi FN, Smith BJ, O'Donnell MA, Konety BR. The impact of age on the response of patients with superficial
AC
112.
bladder cancer to intravesical immunotherapy. J Urol 2006; 175:1634-9; discussion 9-40. 113.
Margel D, Alkhateeb SS, Finelli A, Fleshner N. Diminished efficacy of Bacille Calmette-Guerin among elderly patients with nonmuscle invasive bladder cancer. Urology 2011; 78:848-54.
114.
Herr HW. Age and outcome of superficial bladder cancer treated with bacille Calmette-Guerin therapy. Urology 2007; 70:65-8.
31
ACCEPTED MANUSCRIPT 115.
Hurez V, Padron AS, Svatek RS, Curiel TJ. Considerations for successful cancer immunotherapy in aged hosts. Clin Exp Immunol 2017; 187:53-63.
116.
Begley LA, Kasina S, MacDonald J, Macoska JA. The inflammatory microenvironment of the aging prostate facilitates cellular proliferation and hypertrophy. Cytokine 2008; 43:194-9.
117.
Barron DA, Strand DW, Ressler SJ, Dang TD, Hayward SW, Yang F, Ayala GE, Ittmann M, Rowley DR. TGF-beta1
T
induces an age-dependent inflammation of nerve ganglia and fibroplasia in the prostate gland stroma of a novel
Bianchi-Frias D, Vakar-Lopez F, Coleman IM, Plymate SR, Reed MJ, Nelson PS. The effects of aging on the
CR
118.
IP
transgenic mouse. PLoS One 2010; 5:e13751.
molecular and cellular composition of the prostate microenvironment. PLoS One 2010; 5. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G,
US
119.
AN
Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI. Human gut microbiome viewed across age and geography. Nature
Pitt JM, Vetizou M, Waldschmitt N, Kroemer G, Chamaillard M, Boneca IG, Zitvogel L. Fine-Tuning Cancer
ED
120.
M
2012; 486:222-7.
Immunotherapy: Optimizing the Gut Microbiome. Cancer Res 2016; 76:4602-7. Zitvogel L, Ayyoub M, Routy B, Kroemer G. Microbiome and Anticancer Immunosurveillance. Cell 2016; 165:276-
PT
121.
122.
CE
87.
Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre
AC
ML, Chang EB, Gajewski TF. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PDL1 efficacy. Science 2015; 350:1084-9. 123.
Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, Poirier-Colame V, Roux A, Becharef S, Formenti S, Golden E, Cording S, Eberl G, Schlitzer A, Ginhoux F, Mani S, Yamazaki T, Jacquelot N, Enot DP, Berard M, Nigou J, Opolon P, Eggermont A, Woerther PL, Chachaty E, Chaput N, Robert C, Mateus C, Kroemer G, Raoult D, Boneca IG, Carbonnel F, Chamaillard M, Zitvogel L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350:1079-84. 32
ACCEPTED MANUSCRIPT 124.
Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, Schlitzer A, Ginhoux F, Apetoh L, Chachaty E, Woerther PL, Eberl G, Berard M, Ecobichon C, Clermont D, Bizet C, Gaboriau-Routhiau V, Cerf-Bensussan N, Opolon P, Yessaad N, Vivier E, Ryffel B, Elson CO, Dore J, Kroemer G, Lepage P, Boneca IG, Ghiringhelli F, Zitvogel L. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013; 342:971-6.
T
Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, Dai
IP
RM, Kiu H, Cardone M, Naik S, Patri AK, Wang E, Marincola FM, Frank KM, Belkaid Y, Trinchieri G, Goldszmid RS.
CR
Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science
CE
PT
ED
M
AN
US
2013; 342:967-70.
AC
125.
33
AC
CE
PT
ED
M
AN
US
CR
IP
T
ACCEPTED MANUSCRIPT
Fig. 1. Example of an age-specific immune effect that can be ameliorated to allow effective anti-meanoma immunity. (a) Young mice challenged with B16 melanoma experience increased regulatory T cells (T regs), but not myeloid derived suppressor cells (MDSC), whereas aged mice experience increased Tregs and MDSC. In the aged, the increased MDSC could be due to poor Treg control of them directly, or through indirect mechanisms (denoted by question marks). (b) In young mice, denileukin diftitox (DD) reduces T regs with little MDSC effect, improving antitumour immunity [increased +
interferon (IFN)-γ T cells]. By contrast, in aged mice (c), DD-mediated Treg reduction reduces Tregs but without significant
34
ACCEPTED MANUSCRIPT +
+
increase in IFN-γ T cells. Interleukin (IL)–17 , potentially detrimental T cells, increase, as do deleterious MDSC, thereby inhibiting beneficial anti-tumour immunity. Green anti-tumour immunity means good immunity, whereas red means less effective immunity. (d) By adding anti-granulocyte-differentiation antigen-1 (αGr-1) antibody to DD in aged mice, the MDSC increase is blunted and aged mice now mount anti-tumour immunity comparable to young hosts receiving DD alone, with comparable clinical efficacy. Adding αGr-1 to DD in young hosts does not improve immune or clinical effects
AC
CE
PT
ED
M
AN
US
CR
IP
T
further, as MDSC were not increased further by DD. The red ‘X’ denotes DD effects to reduce Treg inhibition.
35
ACCEPTED MANUSCRIPT Highlights
This article reviews major findings in age effects on the immune system and how they affect the success of cancer immunotherapy. Differences and similarities between mouse models and human data are discussed. This background is
T
used to summarize key findings in efforts to use cancer immunotherapy successfully and optimally in aged patients.
IP
Major reviews are addressed and original source data are discussed. Areas for further investigations and testing are
AC
CE
PT
ED
M
AN
US
CR
discussed.
36